51
|
Hoegen-Saßmannshausen P, Jessen I, Buchele C, Schlüter F, Rippke C, Renkamp CK, Weykamp F, Regnery S, Liermann J, Meixner E, Hoeltgen L, Eichkorn T, König L, Debus J, Klüter S, Hörner-Rieber J. Clinical Outcomes of Online Adaptive Magnetic Resonance-Guided Stereotactic Body Radiotherapy of Adrenal Metastases from a Single Institution. Cancers (Basel) 2024; 16:2273. [PMID: 38927978 PMCID: PMC11201609 DOI: 10.3390/cancers16122273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
(1) Background: Recent publications foster stereotactic body radiotherapy (SBRT) in patients with adrenal oligometastases or oligoprogression. However, local control (LC) after non-adaptive SBRT shows the potential for improvement. Online adaptive MR-guided SBRT (MRgSBRT) improves tumor coverage and organ-at-risk (OAR) sparing. Long-term results of adaptive MRgSBRT are still sparse. (2) Methods: Adaptive MRgSBRT was performed on a 0.35 T MR-Linac. LC, overall survival (OS), progression-free survival (PFS), overall response rate (ORR), and toxicity were assessed. (3) Results: 35 patients with 40 adrenal metastases were analyzed. The median gross tumor volume was 30.6 cc. The most common regimen was 10 fractions at 5 Gy. The median biologically effective dose (BED10) was 75.0 Gy. Plan adaptation was performed in 98% of all fractions. The median follow-up was 7.9 months. One local failure occurred after 16.6 months, resulting in estimated LC rates of 100% at one year and 90% at two years. ORR was 67.5%. The median OS was 22.4 months, and the median PFS was 5.1 months. No toxicity > CTCAE grade 2 occurred. (4) Conclusions: LC and ORR after adrenal adaptive MRgSBRT were excellent, even in a cohort with comparably large metastases. A BED10 of 75 Gy seems sufficient for improved LC in comparison to non-adaptive SBRT.
Collapse
Affiliation(s)
- Philipp Hoegen-Saßmannshausen
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Inga Jessen
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Carolin Buchele
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Fabian Schlüter
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Carolin Rippke
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Claudia Katharina Renkamp
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Fabian Weykamp
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sebastian Regnery
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Jakob Liermann
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Eva Meixner
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Line Hoeltgen
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Tanja Eichkorn
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Laila König
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Sebastian Klüter
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Juliane Hörner-Rieber
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
52
|
Tirtei E, Campello A, Sciannameo V, Asaftei SD, Meazza C, Sironi G, Longhi A, Ibrahim T, Tamburini A, Coccoli L, Crocco F, Cagnazzo C, De Luna E, Quarello P, Berchialla P, Fagioli F. Prolonged 14-day continuous infusion of high-dose ifosfamide for patients with relapsed and refractory high-grade osteosarcoma: a retrospective multicentre cohort study. BMC Cancer 2024; 24:747. [PMID: 38898388 PMCID: PMC11186082 DOI: 10.1186/s12885-024-12498-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The prognosis of patients with Relapsed/Refractory Osteosarcoma (R/R OS) remains dismal without an agreement on systemic therapy. The use of High-Dose Ifosfamide (14 g/sqm) with an external pump in outpatient setting (14-IFO) in R/R OS patients is limited. This study represents the first retrospective cohort analysis focused on evaluating the activity and toxicity of 14-IFO in this setting. PATIENTS AND METHODS The study investigated 14-IFO activity, in terms of tumour response according to RECIST 1.1 criteria, as well as survival rates and toxicity, according to CTCAE v.5. RESULTS The trial enrolled 26 patients with R/R OS. The Overall Response Rate (ORR) and Disease Control Rate (DCR) obtained was 23% and 57.5%, respectively. Patients with relapsed OS showed a higher ORR (45%) and DCR (82%) compared to refractory patients, irrespective of the number of prior treatment lines received. The achievement of disease control with 14-IFO administration enabled 27% of patients to undergo new local treatment. Four-month Progression-Free Survival (PFS) was 54% for all patients and 82% for the relapsed OS sub-group. Median Overall Survival (OSurv) was 13.7 months, with 1-year OSurv of 51% for all patients and 71% for relapsed patients. Age over 18 years and the presence of refractory disease were identified as negative prognostic factors for this patient cohort. A total of 101 cycles were evaluated for toxic assessment, demonstrating a tolerable profile without grade 3-4 non-haematological toxicities. CONCLUSIONS 14-IFO should be considered a viable treatment option for R/R OS, particularly due to its well tolerated toxicity profile and the potential for home-administration, which can improve patient quality of life without compromising efficacy.
Collapse
Affiliation(s)
- Elisa Tirtei
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Anna Campello
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy.
| | - Veronica Sciannameo
- Centre for Biostatistics, Epidemiology and Public Health, Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Italy.
| | - Sebastian Dorin Asaftei
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy
| | - Cristina Meazza
- Paediatric Oncology Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanna Sironi
- Paediatric Oncology Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Longhi
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Angela Tamburini
- Department of Paediatric Haematology-Oncology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Luca Coccoli
- Pediatric Oncology-Hematology Unit, Stem Cell Transplantation and EURACAN Hub Center Unit, S. Chiara Hospital, AOUP, Pisa, Italy
| | - Fanj Crocco
- Paediatrics Division, Department of Health Sciences, AOU Maggiore della Carità di Novara, Piemonte Orientale University, Novara, Italy
| | - Celeste Cagnazzo
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy
| | - Elvira De Luna
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy
| | - Paola Quarello
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Paola Berchialla
- Centre for Biostatistics, Epidemiology and Public Health, Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Franca Fagioli
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, 10126, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| |
Collapse
|
53
|
Velimirovici MD, Feier CVI, Vonica RC, Faur AM, Muntean C. Efficacy and Safety of Atezolizumab as a PD-L1 Inhibitor in the Treatment of Cervical Cancer: A Systematic Review. Biomedicines 2024; 12:1291. [PMID: 38927498 PMCID: PMC11200956 DOI: 10.3390/biomedicines12061291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The efficacy and safety of PD-L1 inhibitors in the treatment of cervical cancer is an ongoing research question. This review aims to establish a clear profile of atezolizumab, examining its impact on survival outcomes, response rates, and safety measured by serious adverse events (SAEs). MATERIALS AND METHODS A literature search was conducted using PubMed, Scopus, and Web of Science, focusing on articles published up to February 2024. The review followed the PRISMA guidelines and synthesized outcomes from four randomized trial studies involving atezolizumab administered at 1200 mg IV every three weeks, alone or in combination with chemoradiotherapy. RESULTS A total of 284 patients received atezolizumab, the majority being advanced stage cervical cancer (IVA-IVB). Median follow-up times ranged from 9 weeks to 32.9 months. It was found that combining atezolizumab with standard therapies extended median progression-free survival (PFS) from 10.4 to 13.7 months and overall survival (OS) from 22.8 to 32.1 months, according to the phase III trial. Monotherapy and initial treatment settings with atezolizumab also showed promising efficacy, with disease-free survival rates at 24 months reaching 79% compared to 52% with standard therapy alone. However, the treatment was associated with high rates of SAEs, reaching up to 79% in more intensive treatment combinations. CONCLUSIONS Atezolizumab demonstrates significant potential in improving PFS and OS in patients with cervical cancer, supporting its inclusion as a first-line treatment option. Despite the efficacy benefits, the high incidence of SAEs necessitates careful patient selection and management strategies to mitigate risks. This systematic review supports the continued evaluation of atezolizumab in broader clinical trials to refine its therapeutic profile and safety measures in the context of cervical cancer treatment.
Collapse
Affiliation(s)
- Milan Daniel Velimirovici
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timisoara, Romania;
| | - Catalin Vladut Ionut Feier
- First Discipline of Surgery, Department X-Surgery, “Victor Babes” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timisoara, Romania;
- First Surgery Clinic, “Pius Brinzeu” Clinical Emergency Hospital, 300723 Timisoara, Romania
| | - Razvan Constantin Vonica
- Preclinical Department, Discipline of Physiology, Faculty of General Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania
| | - Alaviana Monique Faur
- Faculty of Medicine, Victor Babes” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timisoara, Romania;
| | - Calin Muntean
- Medical Informatics and Biostatistics, Department III-Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timisoara, Romania;
| |
Collapse
|
54
|
Mansour N, Heinrich K, Zhang D, Winkelmann M, Ingenerf M, Gold L, Klambauer K, Rudelius M, Klauschen F, von Bergwelt-Baildon M, Ricke J, Heinemann V, Westphalen CB, Kunz WG. Patient eligibility for trials with imaging response assessment at the time of molecular tumor board presentation. Cancer Imaging 2024; 24:70. [PMID: 38849902 PMCID: PMC11157753 DOI: 10.1186/s40644-024-00708-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/11/2024] [Indexed: 06/09/2024] Open
Abstract
PURPOSE To assess the eligibility of patients with advanced or recurrent solid malignancies presented to a molecular tumor board (MTB) at a large precision oncology center for inclusion in trials with the endpoints objective response rate (ORR) or duration of response (DOR) based on Response Evaluation Criteria in Solid Tumors (RECIST version 1.1). METHODS Prospective patients with available imaging at the time of presentation in the MTB were included. Imaging data was reviewed for objectifiable measurable disease (MD) according to RECIST v1.1. Additionally, we evaluated the patients with MD for representativeness of the identified measurable lesion(s) in relation to the overall tumor burden. RESULTS 262 patients with different solid malignancies were included. 177 patients (68%) had MD and 85 (32%) had non-measurable disease (NMD) at the time point of MTB presentation in accordance with RECIST v1.1. MD was not representative of the overall tumor burden in eleven patients (6%). The main reasons for NMD were lesions with longest diameter shorter than 10 mm (22%) and non-measurable peritoneal carcinomatosis (18%). Colorectal cancer and malignant melanoma displayed the highest rates of MD (> 75%). In contrast, gastric cancer, head and neck malignancies, and ovarian carcinoma had the lowest rates of MD (< 55%). In case of MD, the measurable lesions were representative of the overall tumor burden in the vast majority of cases (94%). CONCLUSION Approximately one third of cancer patients with advanced solid malignancies are not eligible for treatment response assessment in trials with endpoints ORR or DOR at the time of MTB presentation. The rate of patients eligible for trials with imaging endpoints differs significantly based on the underlying malignancy and should be taken under consideration during the planning of new precision oncology trials.
Collapse
Affiliation(s)
- Nabeel Mansour
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
| | - Danmei Zhang
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK partner site Munich), Heidelberg, Germany
| | - Michael Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Maria Ingenerf
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lukas Gold
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Konstantin Klambauer
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK partner site Munich), Heidelberg, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany.
| |
Collapse
|
55
|
Zhu C, Zhang C, Wang S, Xun Z, Zhang D, Lan Z, Zhang L, Chao J, Liang Y, Pu Z, Ning C, Sang X, Yang X, Wang H, Jiang X, Zhao H. Characterizations of multi-kingdom gut microbiota in immune checkpoint inhibitor-treated hepatocellular carcinoma. J Immunother Cancer 2024; 12:e008686. [PMID: 38844407 PMCID: PMC11163665 DOI: 10.1136/jitc-2023-008686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND The association between gut bacteria and the response to immune checkpoint inhibitors (ICI) in hepatocellular carcinoma (HCC) has been studied; however, multi-kingdom gut microbiome alterations and interactions in ICI-treated HCC cohorts are not fully understood. METHODS From November 2018 to April 2022, patients receiving ICI treatment for advanced HCC were prospectively enrolled. Herein, we investigated the multi-kingdom microbiota characterization of the gut microbiome, mycobiome, and metabolome using metagenomic, ITS2, and metabolomic data sets of 80 patients with ICI-treated HCC. RESULTS Our findings demonstrated that bacteria and metabolites differed significantly between the durable clinical benefit (DCB) and non-durable clinical benefit (NDB) groups, whereas the differences were smaller for fungi. The overall diversity of bacteria and fungi before treatment was higher in the DCB group than in the NDB group, and the difference in diversity began to change with the use of immunotherapy after 6-8 weeks. We also explored the alterations of gut microbes in the DCB and NDB groups, established 18 bacterial species models as predictive biomarkers for predicting whether immunotherapy is of sustained benefit (area under the curve=75.63%), and screened two species of bacteria (Actinomyces_sp_ICM47, and Senegalimassilia_anaerobia) and one metabolite (galanthaminone) as prognostic biomarkers for predicting survival in patients with HCC treated with ICI. CONCLUSIONS In this study, the status and characterization of the multi-kingdom microbiota, including gut bacteria, fungi, and their metabolites, were described by multiomics sequencing for the first time in patients with HCC treated with ICI. Our findings demonstrate the potential of bacterial taxa as predictive biomarkers of ICI clinical efficacy, and bacteria and their metabolites as prognostic biomarkers.
Collapse
Affiliation(s)
- Chengpei Zhu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
- Department of General Surgery Center, Beijing Youan Hospital, Clinical Center for Liver Cancer, Capital Medical University, Beijing, China
| | - Chenchen Zhang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Ziyu Xun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Dongya Zhang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, China
| | - Zhou Lan
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiashuo Chao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yajun Liang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, China
| | - Zilun Pu
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, China
| | - Cong Ning
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Hanping Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xianzhi Jiang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, China
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| |
Collapse
|
56
|
Zhu C, Wang Y, Zhu R, Wang S, Xue J, Zhang D, Lan Z, Zhang C, Liang Y, Zhang N, Xun Z, Zhang L, Ning C, Yang X, Chao J, Long J, Yang X, Wang H, Sang X, Jiang X, Zhao H. Gut microbiota and metabolites signatures of clinical response in anti-PD-1/PD-L1 based immunotherapy of biliary tract cancer. Biomark Res 2024; 12:56. [PMID: 38831368 PMCID: PMC11149318 DOI: 10.1186/s40364-024-00607-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/30/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Accumulating evidence suggests that the gut microbiota and metabolites can modulate tumor responses to immunotherapy; however, limited data has been reported on biliary tract cancer (BTC). This study used metagenomics and metabolomics to identify characteristics of the gut microbiome and metabolites in immunotherapy-treated BTC and their potential as prognostic and predictive biomarkers. METHODS This prospective cohort study enrolled 88 patients with BTC who received PD-1/PD-L1 inhibitors from November 2018 to May 2022. The microbiota and metabolites significantly enriched in different immunotherapy response groups were identified through metagenomics and LC-MS/MS. Associations between microbiota and metabolites, microbiota and clinical factors, and metabolites and clinical factors were explored. RESULTS Significantly different bacteria and their metabolites were both identified in the durable clinical benefit (DCB) and non-durable clinical benefit (NDB) groups. Of these, 20 bacteria and two metabolites were significantly associated with survival. Alistipes were positively correlated with survival, while Bacilli, Lactobacillales, and Pyrrolidine were negatively correlated with survival. Predictive models based on six bacteria, four metabolites, and the combination of three bacteria and two metabolites could all discriminated between patients in the DCB and NDB groups with high accuracy. Beta diversity between two groups was significantly different, and the composition varied with differences in the use of immunotherapy. CONCLUSIONS Patients with BTC receiving immunotherapy have specific alterations in the interactions between microbiota and metabolites. These findings suggest that gut microbiota and metabolites are potential prognostic and predictive biomarkers for clinical outcomes of anti-PD-1/PD-L1-treated BTC.
Collapse
Affiliation(s)
- Chengpei Zhu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
- Department of General Surgery Center, Clinical Center for Liver Cancer, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yunchao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
- Organ Transplantation Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Ruijuan Zhu
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jingnan Xue
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Dongya Zhang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Zhou Lan
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Chenchen Zhang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Yajun Liang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Ziyu Xun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Cong Ning
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xu Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jiashuo Chao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Hanping Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Xinting Sang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Xianzhi Jiang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| |
Collapse
|
57
|
Hoegen-Saßmannshausen P, Naumann P, Hoffmeister-Wittmann P, Ben Harrabi S, Seidensaal K, Weykamp F, Mielke T, Ellerbrock M, Habermehl D, Springfeld C, Dill MT, Longerich T, Schirmacher P, Mehrabi A, Chang DH, Hörner-Rieber J, Jäkel O, Haberer T, Combs SE, Debus J, Herfarth K, Liermann J. Carbon ion radiotherapy of hepatocellular carcinoma provides excellent local control: The prospective phase I PROMETHEUS trial. JHEP Rep 2024; 6:101063. [PMID: 38737600 PMCID: PMC11087711 DOI: 10.1016/j.jhepr.2024.101063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/22/2024] [Accepted: 03/07/2024] [Indexed: 05/14/2024] Open
Abstract
Background & Aims Inoperable hepatocellular carcinoma (HCC) can be treated by stereotactic body radiotherapy. However, carbon ion radiotherapy (CIRT) is more effective for sparing non-tumorous liver. High linear energy transfer could promote therapy efficacy. Japanese and Chinese studies on hypofractionated CIRT have yielded excellent results. Because of different radiobiological models and the different etiological spectrum of HCC, applicability of these results to European cohorts and centers remains questionable. The aim of this prospective study was to assess safety and efficacy and to determine the optimal dose of CIRT with active raster scanning based on the local effect model (LEM) I. Methods CIRT was performed every other day in four fractions with relative biological effectiveness (RBE)-weighted fraction doses of 8.1-10.5 Gy (total doses 32.4-42.0 Gy [RBE]). Dose escalation was performed in five dose levels with at least three patients each. The primary endpoint was acute toxicity after 4 weeks. Results Twenty patients received CIRT (median age 74.7 years, n = 16 with liver cirrhosis, Child-Pugh scores [CP] A5 [n = 10], A6 [n = 4], B8 [n = 1], and B9 [n = 1]). Median follow up was 23 months. No dose-limiting toxicities and no toxicities exceeding grade II occurred, except one grade III gamma-glutamyltransferase elevation 12 months after CIRT, synchronous to out-of-field hepatic progression. During 12 months after CIRT, no CP elevation occurred. The highest dose level could be applied safely. No local recurrence developed during follow up. The objective response rate was 80%. Median overall survival was 30.8 months (1/2/3 years: 75%/64%/22%). Median progression-free survival was 20.9 months (1/2/3 years: 59%/43%/43%). Intrahepatic progression outside of the CIRT target volume was the most frequent pattern of progression. Conclusions CIRT of HCC yields excellent local control without dose-limiting toxicity. Impact and implications To date, safety and efficacy of carbon ion radiotherapy for hepatocellular carcinoma have only been evaluated prospectively in Japanese and Chinese studies. The optimal dose and fractionation when using the local effect model for radiotherapy planning are unknown. The results are of particular interest for European and American particle therapy centers, but also of relevance for all specialists involved in the treatment and care of patients with hepatocellular carcinoma, as we present the first prospective data on carbon ion radiotherapy in hepatocellular carcinoma outside of Asia. The excellent local control should encourage further use of carbon ion radiotherapy for hepatocellular carcinoma and design of randomized controlled trials. Clinical Trials Registration The study is registered at ClinicalTrials.gov (NCT01167374).
Collapse
Affiliation(s)
- Philipp Hoegen-Saßmannshausen
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Naumann
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Xcare Praxis für Strahlentherapie, Saarbrücken, Germany
| | - Paula Hoffmeister-Wittmann
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Semi Ben Harrabi
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina Seidensaal
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Fabian Weykamp
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Mielke
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Malte Ellerbrock
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Habermehl
- Wilhelm-Conrad-Röntgen-Klinik Gießen, Universitätsklinikum Gießen und Marburg GmbH, Gießen, Germany
| | - Christoph Springfeld
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Medical Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
| | - Michael T. Dill
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
- Department of Gastroenterology, Infectious Diseases, Intoxication, Heidelberg University Hospital, Heidelberg, Germany
- Experimental Hepatology, Inflammation and Cancer Research Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Thomas Longerich
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Schirmacher
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Arianeb Mehrabi
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
- Department of General, Visceral & Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - De-Hua Chang
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Juliane Hörner-Rieber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Jäkel
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- Medical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technical University of Munich (TUM), Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, Neuherberg, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, Heidelberg, Germany
| | - Klaus Herfarth
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Jakob Liermann
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- Liver Cancer Centre Heidelberg, Heidelberg, Germany
| |
Collapse
|
58
|
Liu H, Zhu C, Wang X, Chen X, Li Z, Xian J. Prediction of pathological complete response in locally advanced head and neck squamous cell carcinoma treated with neoadjuvant chemo-immunotherapy using volumetric multisequence MRI histogram analysis. Neuroradiology 2024; 66:919-929. [PMID: 38503986 DOI: 10.1007/s00234-024-03339-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE This study aimed to develop a multisequence MRI-based volumetric histogram metrics model for predicting pathological complete response (pCR) in advanced head and neck squamous cell carcinoma (HNSCC) patients undergoing neoadjuvant chemo-immunotherapy (NCIT) and compare its predictive performance with AJCC staging and RECIST 1.1 criteria. METHODS Twenty-four patients with locally advanced HNSCC from a prospective phase II trial were enrolled for analysis. All patients underwent pre- and post-NCIT MRI examinations from which whole-tumor histogram features were extracted, including T1WI, T2WI, enhanced T1WI (T1Gd), diffusion-weighted imaging (DWI) sequences, and their corresponding apparent diffusion coefficient (ADC) maps. The pathological results divided the patients into pathological complete response (pCR) and non-pCR (N-pCR) groups. Delta features were calculated as the percentage change in histogram features from pre- to post-treatment. After data reduction and feature selection, logistic regression was used to build prediction models. ROC analysis was performed to assess the diagnostic performance. RESULTS Eleven of 24 patients achieved pCR. Pre_T2_original_firstorder_Minimum, Post_ADC_original_firstorder_MeanAbsoluteDeviation, and Delta_T1Gd_original_firstorder_Skewness were associated with achieving pCR after NCIT. The Combined_Model demonstrated the best predictive performance (AUC 0.95), outperforming AJCC staging (AUC 0.52) and RECIST 1.1 (AUC 0.72). The Pre_Model (AUC 0.83) or Post-Model (AUC 0.83) had a better predictive ability than AJCC staging. CONCLUSION Multisequence MRI-based volumetric histogram analysis can non-invasively predict the pCR status of HNSCC patients undergoing NCIT. The use of histogram features extracted from pre- and post-treatment MRI exhibits promising predictive performance and offers a novel quantitative assessment method for evaluating pCR in HNSCC patients receiving NCIT.
Collapse
Affiliation(s)
- Hangzhi Liu
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, NO.1 of Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China
| | - Changyu Zhu
- Cancer Center, Beijing Tongren Hospital, Capital Medical University, NO.1 of Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China
| | - Xinyan Wang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, NO.1 of Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China
| | - Xiaohong Chen
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, No.1 of Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China
| | - Zhixin Li
- Cancer Center, Beijing Tongren Hospital, Capital Medical University, NO.1 of Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, NO.1 of Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
59
|
Cerou M, Thai H, Deyme L, Fliscounakis‐Huynh S, Comets E, Cohen P, Cartot‐Cotton S, Veyrat‐Follet C. Joint modeling of tumor dynamics and progression-free survival in advanced breast cancer: Leveraging data from amcenestrant early phase I-II trials. CPT Pharmacometrics Syst Pharmacol 2024; 13:941-953. [PMID: 38558299 PMCID: PMC11179707 DOI: 10.1002/psp4.13128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
A joint modeling framework was developed using data from 75 patients of early amcenestrant phase I-II AMEERA-1-2 dose escalation and expansion cohorts. A semi-mechanistic tumor growth inhibition (TGI) model was developed. It accounts for the dynamics of sensitive and resistant tumor cells, an exposure-driven effect on tumor proliferation of sensitive cells, and a delay in the initiation of treatment effect to describe the time course of target lesion tumor size (TS) data. Individual treatment exposure overtime was introduced in the model using concentrations predicted by a population pharmacokinetic model of amcenestrant. This joint modeling framework integrated complex RECISTv1.1 criteria information, linked TS metrics to progression-free survival (PFS), and was externally evaluated using the randomized phase II trial AMEERA-3. We demonstrated that the instantaneous rate of change in TS (TS slope) was an important predictor of PFS and the developed joint model was able to predict well the PFS of amcenestrant phase II monotherapy trial using only early phase I-II data. This provides a good modeling and simulation tool to inform early development decisions.
Collapse
Affiliation(s)
- Marc Cerou
- Data and Data Science, Translational Disease Modeling OncologySanofi R&DParisFrance
| | - Hoai‐Thu Thai
- Data and Data Science, Translational Disease Modeling OncologySanofi R&DParisFrance
| | - Laure Deyme
- Translational Medicine & Early Development, Modeling & SimulationSanofi R&DMontpellierFrance
| | | | - Emmanuelle Comets
- IAME, InsermUniversité Paris CitéParisFrance
- Irset (Institut de Recherche en Santé, Environnement et Travail) ‐ UMR_S 1085Univ Rennes, Inserm, EHESPRennesFrance
| | | | - Sylvaine Cartot‐Cotton
- Pharmacokinetics Dynamics and Metabolism, Translational Medicine & Early DevelopmentSanofi R&DChilly MazarinFrance
| | | |
Collapse
|
60
|
Kodama T, Imajima T, Shimokawa M, Otsuka T, Kawahira M, Nakazawa J, Hori T, Shibuki T, Arima S, Ido A, Miwa K, Okabe Y, Koga F, Ueda Y, Kubotsu Y, Shimokawa H, Takeshita S, Nishikawa K, Komori A, Otsu S, Hosokawa A, Sakai T, Sakai K, Oda H, Kawahira M, Arita S, Honda T, Taguchi H, Tsuneyoshi K, Kawaguchi Y, Fujita T, Sakae T, Shirakawa T, Mizuta T, Mitsugi K. A multicenter retrospective observational NAPOLEON2 study of nanoliposomal irinotecan with fluorouracil and folinic acid in patients with unresectable pancreatic cancer. Sci Rep 2024; 14:12422. [PMID: 38816500 PMCID: PMC11139902 DOI: 10.1038/s41598-024-63172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024] Open
Abstract
Nanoliposomal irinotecan with fluorouracil and folinic acid (NFF) is a standard regimen after gemcitabine-based therapy for patients with unresectable or recurrent pancreatic cancer. However, there are limited clinical data on its efficacy and safety in the real-world. We therefore initiated a retrospective and prospective observational study (NAPOLEON-2). The results of the retrospective part were reported herein. In this retrospective study, we evaluated 161 consecutive patients who received NFF as second-or-later-line regimen. The main endpoint was overall survival (OS), and the other endpoints were response rate, disease control rate, progression-free survival (PFS), dose intensity, and adverse events (AEs). The median age was 67 years (range, 38-85 years). The median OS and PFS were 8.1 and 3.4 months, respectively. The objective response and disease control rates were 5% and 52%, respectively. The median relative dose intensity was 81.6% for nanoliposomal irinotecan and 82.9% for fluorouracil. Grade 3 or 4 hematological and nonhematological AEs occurred in 47 and 42 patients, respectively. Common grade 3 or 4 AEs included neutropenia (24%), anorexia (12%), and leukocytopenia (12%). Subanalysis of patients treated with second-line and third-or-later-line demonstrated no statistical significant difference in OS (7.6 months vs. 9.1 months, respectively; hazard ratio, 0.92; 95% confidence interval, 0.64-1.35; p = 0.68). In conclusion, NFF has acceptable efficacy and safety profile even in real-world clinical settings. The prospective study is in progress to validate these findings.
Collapse
Affiliation(s)
- Tomoko Kodama
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Takashi Imajima
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-Cho, Sasebo-Shi, Nagasaki, 857-8575, Japan
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi‑ku, Fukuoka-Shi, Fukuoka, 812-8582, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, 3-1-1 Notame, Minami-Ku, Fukuoka-Shi, Fukuoka, 811-1395, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube-Shi, Yamaguchi, 755-8505, Japan
| | - Taiga Otsuka
- Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-Ku, Fukuoka-Shi, Fukuoka, 810-0072, Japan
| | - Masahiro Kawahira
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Takeshi Hori
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Taro Shibuki
- Department for the Promotion of Drug and Diagnostic Development, Division of Drug and Diagnostic Development Promotion, Translational Research Support Office, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-Shi, Chiba, 277-8577, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-Shi, Chiba, 277-8577, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-Shi, Kagoshima, 890-8520, Japan
| | - Akio Ido
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-Shi, Kagoshima, 890-8520, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
| | - Yoshinobu Okabe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-Machi, Saga-Shi, Saga, 840-8571, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, 2-1-1 Nagamine-Minami, Higashi-Ku, Kumamoto-Shi, Kumamoto, 861-8520, Japan
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada, Karatsu-Shi, Saga, 847-8588, Japan
| | - Hozumi Shimokawa
- Department of Hematology and Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-Ku, Kitakyushu-Shi, Fukuoka, 806-8501, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, 3-15 Morimachi, Nagasaki-Shi, Nagasaki, 852-8511, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, 160 Kou, Minamiumemoto-Machi, Matsuyama-Shi, Ehime, 791-0280, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, 5200 Kiyotakechoukihara, Miyazaki-Shi, Miyazaki, 889-1692, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, 1-5 Ninomaru, Chuo-Ku, Kumamoto-Shi, Kumamoto, 860-0008, Japan
| | - Kenji Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, 1-5 Ninomaru, Chuo-Ku, Kumamoto-Shi, Kumamoto, 860-0008, Japan
- Department of Clinical Oncology, Japan Community Health Care Organization Hitoyoshi Medical Center, 35 Oikamimachi, Hitoyoshi-Shi, Kumamoto, 868-8555, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, 5-3-1 Chikami, Minami-Ku, Kumamoto-Shi, Kumamoto, 861-4193, Japan
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, 1-13-1 Yojirou, Kagoshima-Shi, Kagoshima, 890-0062, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, 5-30 Kita-Takamatsucho, Miyazaki, 880-8510, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-Shi, Nagasaki, 852-8501, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Izumi General Medical Center, 520 Myoujin-Cho, Izumi-Shi, Kagoshima, 899-0131, Japan
- Department of Gastroenterology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, 520 Myoujin-Cho, Izumi-Shi, Kagoshima, 899-0131, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, 422-1 Raiha, Asakura-Shi, Fukuoka, 838-0069, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-Cho, Satsumasendai-Shi, Kagoshima, 895-0074, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-Cho, Satsumasendai-Shi, Kagoshima, 895-0074, Japan
| | - Tsuyoshi Shirakawa
- Department of Medical Checkup Center, Eikoh Hospital, 3-8-15 Befu-Nishi, Shime-Machi, Kasuya-Gun, Fukuoka, 811-2232, Japan.
- Clinical Hematology Oncology Treatment Study Group, 1-14-6 Muromi-Gaoka, Nishi-Ku, Fukuoka-Shi, Fukuoka, 819-0030, Japan.
| | - Toshihiko Mizuta
- Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga-Shi, Saga, 840-0831, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-Cho, Sasebo-Shi, Nagasaki, 857-8575, Japan
| |
Collapse
|
61
|
Han Z, Zhang Z, Yang X, Li Z, Sang S, Islam MT, Guo AA, Li Z, Wang X, Wang J, Zhang T, Sun Z, Yu L, Wang W, Xiong W, Li G, Jiang Y. Development and interpretation of a pathomics-driven ensemble model for predicting the response to immunotherapy in gastric cancer. J Immunother Cancer 2024; 12:e008927. [PMID: 38749538 PMCID: PMC11097892 DOI: 10.1136/jitc-2024-008927] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Only a subset of patients with gastric cancer experience long-term benefits from immune checkpoint inhibitors (ICIs). Currently, there is a deficiency in precise predictive biomarkers for ICI efficacy. The aim of this study was to develop and validate a pathomics-driven ensemble model for predicting the response to ICIs in gastric cancer, using H&E-stained whole slide images (WSI). METHODS This multicenter study retrospectively collected and analyzed H&E-stained WSIs and clinical data from 584 patients with gastric cancer. An ensemble model, integrating four classifiers: least absolute shrinkage and selection operator, k-nearest neighbors, decision trees, and random forests, was developed and validated using pathomics features, with the objective of predicting the therapeutic efficacy of immune checkpoint inhibition. Model performance was evaluated using metrics including the area under the curve (AUC), sensitivity, and specificity. Additionally, SHAP (SHapley Additive exPlanations) analysis was used to explain the model's predicted values as the sum of the attribution values for each input feature. Pathogenomics analysis was employed to explain the molecular mechanisms underlying the model's predictions. RESULTS Our pathomics-driven ensemble model effectively stratified the response to ICIs in training cohort (AUC 0.985 (95% CI 0.971 to 0.999)), which was further validated in internal validation cohort (AUC 0.921 (95% CI 0.839 to 0.999)), as well as in external validation cohort 1 (AUC 0.914 (95% CI 0.837 to 0.990)), and external validation cohort 2 (0.927 (95% CI 0.802 to 0.999)). The univariate Cox regression analysis revealed that the prediction signature of pathomics-driven ensemble model was a prognostic factor for progression-free survival in patients with gastric cancer who underwent immunotherapy (p<0.001, HR 0.35 (95% CI 0.24 to 0.50)), and remained an independent predictor after multivariable Cox regression adjusted for clinicopathological variables, (including sex, age, carcinoembryonic antigen, carbohydrate antigen 19-9, therapy regime, line of therapy, differentiation, location and programmed death ligand 1 (PD-L1) expression in all patients (p<0.001, HR 0.34 (95% CI 0.24 to 0.50)). Pathogenomics analysis suggested that the ensemble model is driven by molecular-level immune, cancer, metabolism-related pathways, and was correlated with the immune-related characteristics, including immune score, Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data score, and tumor purity. CONCLUSIONS Our pathomics-driven ensemble model exhibited high accuracy and robustness in predicting the response to ICIs using WSIs. Therefore, it could serve as a novel and valuable tool to facilitate precision immunotherapy.
Collapse
Affiliation(s)
- Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
| | - Zhicheng Zhang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China
- JancsiLab, JancsiTech, Hongkong, China
| | - Xianqi Yang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhe Li
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Shengtian Sang
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | - Md Tauhidul Islam
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Alyssa A Guo
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Zihan Li
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Xiaoyan Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
| | - Lequan Yu
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wei Wang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
- School of Clinical Medicine, Tsinghua University, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
62
|
Zhang Z, Zhang J, Cai M, Huang X, Guo X, Zhu D, Guo T, Yu Y. The fibrosis-4 index is a prognostic factor for cholangiocarcinoma patients who received immunotherapy. Front Immunol 2024; 15:1376590. [PMID: 38799431 PMCID: PMC11116781 DOI: 10.3389/fimmu.2024.1376590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Background Research of immunotherapy for cholangiocarcinoma has yielded some results, but more clinical data are needed to prove its efficacy and safety. Moreover, there is a need to identify accessible indexes for selecting patients who may benefit from such treatments. Methods The medical records of 66 cholangiocarcinoma patients who underwent immunotherapy were retrospectively collected. The effectiveness of immunotherapy was assessed by tumor response, progression-free survival (PFS), and overall survival (OS), while safety was evaluated by adverse events during treatment. Univariate and multivariate Cox regression analyses were performed to identify prognostic risk factors for PFS and OS, and Kaplan-Meier curves of potential prognostic factors were drawn. Results Overall, in this study, immunotherapy achieved an objective response rate of 24.2% and a disease control rate of 89.4% for the included patients. The median PFS was 445 days, and the median OS was 772.5 days. Of the 66 patients, 65 experienced adverse events during treatment, but none had severe consequences. Multivariate Cox analysis indicated that tumor number is a prognostic risk factor for disease progression following immunotherapy in cholangiocarcinoma patients, while tumor differentiation and the fibrosis-4 (FIB-4) index are independent risk factors for OS. Conclusion In general, immunotherapy for cholangiocarcinoma is safe, with adverse events remaining within manageable limits, and it can effectively control disease progression in most patients. The FIB-4 index may reflect the potential benefit of immunotherapy for patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Jingzhao Zhang
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Ming Cai
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Xiaorui Huang
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Xinyi Guo
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Dengsheng Zhu
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Tong Guo
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| | - Yahong Yu
- Department of Biliopancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Wuhan, Hubei, China
| |
Collapse
|
63
|
Wei R, Song J, Pan H, Liu X, Gao J. CPT1C-positive cancer-associated fibroblast facilitates immunosuppression through promoting IL-6-induced M2-like phenotype of macrophage. Oncoimmunology 2024; 13:2352179. [PMID: 38746869 PMCID: PMC11093039 DOI: 10.1080/2162402x.2024.2352179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) exhibit remarkable phenotypic heterogeneity, with specific subsets implicated in immunosuppression in various malignancies. However, whether and how they attenuate anti-tumor immunity in gastric cancer (GC) remains elusive. CPT1C, a unique isoform of carnitine palmitoyltransferase pivotal in regulating fatty acid oxidation, is briefly indicated as a protumoral metabolic mediator in the tumor microenvironment (TME) of GC. In the present study, we initially identified specific subsets of fibroblasts exclusively overexpressing CPT1C, hereby termed them as CPT1C+CAFs. Subsequent findings indicated that CPT1C+CAFs fostered a stroma-enriched and immunosuppressive TME as they correlated with extracellular matrix-related molecular features and enrichment of both immunosuppressive subsets, especially M2-like macrophages, and multiple immune-related pathways. Next, we identified that CPT1C+CAFs promoted the M2-like phenotype of macrophage in vitro. Bioinformatic analyses unveiled the robust IL-6 signaling between CPT1C+CAFs and M2-like phenotype of macrophage and identified CPT1C+CAFs as the primary source of IL-6. Meanwhile, suppressing CPT1C expression in CAFs significantly decreased IL-6 secretion in vitro. Lastly, we demonstrated the association of CPT1C+CAFs with therapeutic resistance. Notably, GC patients with high CPT1C+CAFs infiltration responded poorly to immunotherapy in clinical cohort. Collectively, our data not only present the novel identification of CPT1C+CAFs as immunosuppressive subsets in TME of GC, but also reveal the underlying mechanism that CPT1C+CAFs impair tumor immunity by secreting IL-6 to induce the immunosuppressive M2-like phenotype of macrophage in GC.
Collapse
Affiliation(s)
- Rongyuan Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junquan Song
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongda Pan
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianpeng Gao
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
64
|
Castagnoli F, Mencel J, Ap Dafydd D, Gough J, Drake B, Mcaddy NC, Withey SJ, Riddell AM, Koh DM, Shur JD. Response Evaluation Criteria in Gastrointestinal and Abdominal Cancers: Which to Use and How to Measure. Radiographics 2024; 44:e230047. [PMID: 38662587 DOI: 10.1148/rg.230047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
As the management of gastrointestinal malignancy has evolved, tumor response assessment has expanded from size-based assessments to those that include tumor enhancement, in addition to functional data such as those derived from PET and diffusion-weighted imaging. Accurate interpretation of tumor response therefore requires knowledge of imaging modalities used in gastrointestinal malignancy, anticancer therapies, and tumor biology. Targeted therapies such as immunotherapy pose additional considerations due to unique imaging response patterns and drug toxicity; as a consequence, immunotherapy response criteria have been developed. Some gastrointestinal malignancies require assessment with tumor-specific criteria when assessing response, often to guide clinical management (such as watchful waiting in rectal cancer or suitability for surgery in pancreatic cancer). Moreover, anatomic measurements can underestimate therapeutic response when applied to molecular-targeted therapies or locoregional therapies in hypervascular malignancies such as hepatocellular carcinoma. In these cases, responding tumors may exhibit morphologic changes including cystic degeneration, necrosis, and hemorrhage, often without significant reduction in size. Awareness of pitfalls when interpreting gastrointestinal tumor response is required to correctly interpret response assessment imaging and guide appropriate oncologic management. Data-driven image analyses such as radiomics have been investigated in a variety of gastrointestinal tumors, such as identifying those more likely to respond to therapy or recur, with the aim of delivering precision medicine. Multimedia-enhanced radiology reports can facilitate communication of gastrointestinal tumor response by automatically embedding response categories, key data, and representative images. ©RSNA, 2024 Test Your Knowledge questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Francesca Castagnoli
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Justin Mencel
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Derfel Ap Dafydd
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Jessica Gough
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Brent Drake
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Naami Charlotte Mcaddy
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Samuel Joseph Withey
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Angela Mary Riddell
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Dow-Mu Koh
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Joshua David Shur
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| |
Collapse
|
65
|
Pelc Z, Sędłak K, Leśniewska M, Mielniczek K, Chawrylak K, Skórzewska M, Ciszewski T, Czechowska J, Kiszczyńska A, Wijnhoven BPL, Van Sandick JW, Gockel I, Gisbertz SS, Piessen G, Eveno C, Bencivenga M, De Manzoni G, Baiocchi GL, Morgagni P, Rosati R, Fumagalli Romario U, Davies A, Endo Y, Pawlik TM, Roviello F, Bruns C, Polkowski WP, Rawicz-Pruszyński K. Textbook Neoadjuvant Outcome-Novel Composite Measure of Oncological Outcomes among Gastric Cancer Patients Undergoing Multimodal Treatment. Cancers (Basel) 2024; 16:1721. [PMID: 38730672 PMCID: PMC11083243 DOI: 10.3390/cancers16091721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
The incidence of gastric cancer (GC) is expected to increase to 1.77 million cases by 2040. To improve treatment outcomes, GC patients are increasingly treated with neoadjuvant chemotherapy (NAC) prior to curative-intent resection. Although NAC enhances locoregional control and comprehensive patient care, survival rates remain poor, and further investigations should establish outcomes assessment of current clinical pathways. Individually assessed parameters have served as benchmarks for treatment quality in the past decades. The Outcome4Medicine Consensus Conference underscores the inadequacy of isolated metrics, leading to increased recognition and adoption of composite measures. One of the most simple and comprehensive is the "All or None" method, which refers to an approach where a specific set of criteria must be fulfilled for an individual to achieve the overall measure. This narrative review aims to present the rationale for the implementation of a novel composite measure, Textbook Neoadjuvant Outcome (TNO). TNO integrates five objective and well-established components: Treatment Toxicity, Laboratory Tests, Imaging, Time to Surgery, and Nutrition. It represents a desired, multidisciplinary care and hospitalization of GC patients undergoing NAC to identify the treatment- and patient-related data required to establish high-quality oncological care further. A key strength of this narrative review is the clinical feasibility and research background supporting the implementation of the first and novel composite measure representing the "ideal" and holistic care among patients with locally advanced esophago-gastric junction (EGJ) and GC in the preoperative period after NAC. Further analysis will correlate clinical outcomes with the prognostic factors evaluated within the TNO framework.
Collapse
Affiliation(s)
- Zuzanna Pelc
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Katarzyna Sędłak
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Magdalena Leśniewska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Katarzyna Mielniczek
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Katarzyna Chawrylak
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Magdalena Skórzewska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Tomasz Ciszewski
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Joanna Czechowska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Agata Kiszczyńska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Bas P. L. Wijnhoven
- Department of General Surgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Johanna W. Van Sandick
- Department of Surgical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Suzanne S. Gisbertz
- Department of Surgery, Amsterdam UMC location University of Amsterdam, 1007 MB Amsterdam, The Netherlands;
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, University Lille, and Claude Huriez University Hospital, 59000 Lille, France; (G.P.); (C.E.)
| | - Clarisse Eveno
- Department of Digestive and Oncological Surgery, University Lille, and Claude Huriez University Hospital, 59000 Lille, France; (G.P.); (C.E.)
| | - Maria Bencivenga
- Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Giovanni De Manzoni
- Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Gian Luca Baiocchi
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, and Third Division of General Surgery, Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Paolo Morgagni
- Department of General Surgery, Morgagni-Pierantoni Hospital, 47121 Forlì, Italy;
| | - Riccardo Rosati
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Hospital, Vita Salute University, 20132 Milan, Italy;
| | | | - Andrew Davies
- Department of Upper Gastrointestinal and General Surgery, Guy’s and St Thomas’ Hospital, London SE1 7EH, UK;
| | - Yutaka Endo
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.E.); (T.M.P.)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.E.); (T.M.P.)
| | - Franco Roviello
- Department of Medicine, Surgery, and Neurosciences, University of Siena, 53100 Siena, Italy;
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
| | - Wojciech P. Polkowski
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Karol Rawicz-Pruszyński
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| |
Collapse
|
66
|
Cao T, Zhang W, Wang Q, Wang C, Ma W, Zhang C, Ge M, Tian M, Yu J, Jiao A, Wang L, Liu M, Wang P, Guo Z, Zhou Y, Chen S, Yin W, Yi J, Guo H, Han H, Zhang B, Wu K, Fan D, Wang X, Nie Y, Lu Y, Zhao X. Cancer SLC6A6-mediated taurine uptake transactivates immune checkpoint genes and induces exhaustion in CD8 + T cells. Cell 2024; 187:2288-2304.e27. [PMID: 38565142 DOI: 10.1016/j.cell.2024.03.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 12/12/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
Taurine is used to bolster immunity, but its effects on antitumor immunity are unclear. Here, we report that cancer-related taurine consumption causes T cell exhaustion and tumor progression. The taurine transporter SLC6A6 is correlated with aggressiveness and poor outcomes in multiple cancers. SLC6A6-mediated taurine uptake promotes the malignant behaviors of tumor cells but also increases the survival and effector function of CD8+ T cells. Tumor cells outcompete CD8+ T cells for taurine by overexpressing SLC6A6, which induces T cell death and malfunction, thereby fueling tumor progression. Mechanistically, taurine deficiency in CD8+ T cells increases ER stress, promoting ATF4 transcription in a PERK-JAK1-STAT3 signaling-dependent manner. Increased ATF4 transactivates multiple immune checkpoint genes and induces T cell exhaustion. In gastric cancer, we identify a chemotherapy-induced SP1-SLC6A6 regulatory axis. Our findings suggest that tumoral-SLC6A6-mediated taurine deficiency promotes immune evasion and that taurine supplementation reinvigorates exhausted CD8+ T cells and increases the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Tianyu Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wenyao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qi Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China; College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Chen Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China; College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Wanqi Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Minghui Ge
- Simcere Diagnostics Co., Ltd., Nanjing, Jiangsu 210042, China
| | - Miaomiao Tian
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jia Yu
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Liang Wang
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Manjiao Liu
- Simcere Diagnostics Co., Ltd., Nanjing, Jiangsu 210042, China
| | - Pei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhiyu Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yun Zhou
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Shuyi Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wen Yin
- Department of Blood Transfusion, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing Yi
- Department of Blood Transfusion, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hao Guo
- Simcere Diagnostics Co., Ltd., Nanjing, Jiangsu 210042, China
| | - Hua Han
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xin Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
67
|
Donners R, Candito A, Rata M, Sharp A, Messiou C, Koh DM, Tunariu N, Blackledge MD. Inter- and Intra-Patient Repeatability of Radiomic Features from Multiparametric Whole-Body MRI in Patients with Metastatic Prostate Cancer. Cancers (Basel) 2024; 16:1647. [PMID: 38730599 PMCID: PMC11083580 DOI: 10.3390/cancers16091647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Background: We assessed the test-re-test repeatability of radiomics in metastatic castration-resistant prostate cancer (mCPRC) bone disease on whole-body diffusion-weighted (DWI) and T1-weighted Dixon MRI. (2) Methods: In 10 mCRPC patients, 1.5 T MRI, including DWI and T1-weighted gradient-echo Dixon sequences, was performed twice on the same day. Apparent diffusion coefficient (ADC) and relative fat-fraction-percentage (rFF%) maps were calculated. Per study, up to 10 target bone metastases were manually delineated on DWI and Dixon images. All 106 radiomic features included in the Pyradiomics toolbox were derived for each target volume from the ADC and rFF% maps. To account for inter- and intra-patient measurement repeatability, the log-transformed individual target measurements were fitted to a hierarchical model, represented as a Bayesian network. Repeatability measurements, including the intraclass correlation coefficient (ICC), were derived. Feature ICCs were compared with mean ADC and rFF ICCs. (3) Results: A total of 65 DWI and 47 rFF% targets were analysed. There was no significant bias for any features. Pairwise correlation revealed fifteen ADC and fourteen rFF% feature sub-groups, without specific patterns between feature classes. The median intra-patient ICC was generally higher than the inter-patient ICC. Features that describe extremes in voxel values (minimum, maximum, range, skewness, and kurtosis) showed generally lower ICCs. Several mostly shape-based texture features were identified, which showed high inter- and intra-patient ICCs when compared with the mean ADC or mean rFF%, respectively. (4) Conclusions: Pyradiomics texture features of mCRPC bone metastases varied greatly in inter- and intra-patient repeatability. Several features demonstrated good repeatability, allowing for further exploration as diagnostic parameters in mCRPC bone disease.
Collapse
Affiliation(s)
- Ricardo Donners
- University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Antonio Candito
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| | - Mihaela Rata
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| | - Adam Sharp
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| | - Christina Messiou
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| | - Dow-Mu Koh
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| | - Nina Tunariu
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| | - Matthew D. Blackledge
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK; (A.C.); (M.R.); (A.S.); (C.M.); (D.-M.K.); (N.T.)
| |
Collapse
|
68
|
Harvey RD, Miller TM, Hurley PA, Thota R, Black LJ, Bruinooge SS, Boehmer LM, Fleury ME, Kamboj J, Rizvi MA, Symington BE, Tap WD, Waterhouse DM, Levit LA, Merrill JK, Prindiville SA, Pollastro T, Brewer JR, Byatt LP, Hamroun L, Kim ES, Holland N, Nowakowski GS. A call to action to advance patient-focused and decentralized clinical trials. Cancer 2024; 130:1193-1203. [PMID: 38193828 DOI: 10.1002/cncr.35145] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
This commentary is a call to action for a concerted commitment and effort to transform clinical trials and enable people with cancer to participate in clinical trials closer to home. Three key strategies are identified to address major barriers: confront challenges with the interpretation of US Food and Drug Administration Form 1572 requirements (Statement of Investigator); broaden acceptance of local laboratories and imaging centers; and invest in the creation of effective, sustainable partnerships between research centers and local providers.
Collapse
Affiliation(s)
- R Donald Harvey
- Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Therica M Miller
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, New York, USA
| | | | - Ramya Thota
- Intermountain Health, Salt Lake City, Utah, USA
| | | | | | - Leigh M Boehmer
- Association of Community Cancer Centers, Rockville, Maryland, USA
| | - Mark E Fleury
- American Cancer Society Cancer Action Network, Washington, District of Columbia, USA
| | | | | | | | - William D Tap
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Laura A Levit
- American Society of Clinical Oncology, Alexandria, Virginia, USA
| | | | - Sheila A Prindiville
- National Cancer Institute Coordinating Center for Clinical Trials, Bethesda, Maryland, USA
| | - Teri Pollastro
- Metastatic Breast Cancer Alliance, Mercer Island, Washington, USA
| | - Jamie R Brewer
- US Food and Drug Administration, Rockville, Maryland, USA
| | - Leslie P Byatt
- New Mexico Cancer Care Alliance, Albuquerque, New Mexico, USA
| | | | | | - Nicole Holland
- American Society of Clinical Oncology, Alexandria, Virginia, USA
| | | |
Collapse
|
69
|
He Q, Shi X, Yan J, Wu M, Gu C, Yu X. Circulating tumor DNA serial monitoring of relapse and responses to tislelizumab immunotherapy as second‑line monotherapy for metastatic esophageal squamous cell carcinoma: A prospective study. Mol Clin Oncol 2024; 20:29. [PMID: 38414510 PMCID: PMC10895470 DOI: 10.3892/mco.2024.2727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024] Open
Abstract
Anti-programmed cell death 1 immuno-monotherapy has become the second-line standard treatment for advanced esophageal squamous cell carcinoma (ESCC) after the failure of first-line chemotherapy. However, new biomarkers are still needed to identify patients at risk of tumor progression and to select patients with advanced ESCC who are likely to benefit from immunotherapy. A total of 12 patients with advanced ESCC treated with tislelizumab were prospectively enrolled and endoscopic biopsy samples were collected. Plasma was obtained prior to and after every 2-3 treatment cycles with tislelizumab and when disease progression occurred. Targeted sequencing of 425 genes from plasma cell-free DNA, DNA from leukocytes and fixed esophageal tumor biopsies was performed. The patients underwent imaging analyses every 6-8 weeks until disease progression. The association between status of circulating tumor DNA (ctDNA) or changes in ctDNA following tislelizumab immunotherapy and response, tumor progression and survival was determined. All patients had evaluable next-generation sequencing results at the time of analysis. The results showed that patients with ESCC with liver metastasis had a significantly shorter median progression-free survival (mPFS: 1.4 vs. 11.7 months; P=0.037). TSC complex subunit 2 [11.7 months vs. not reached (NR); P=0.004] and zinc finger protein 217 (11.7 months vs. NR; P=0.022) gene mutations were the independent and negative prognostic factors for median overall survival (OS), respectively. Of note, ctDNA dynamic changes expressed as ∆ mutant molecules per milliliter of plasma (∆MMPM; MMPM detected at the first monitoring time-point after the first infusion of tislelizumab as baseline MMPM) predicted progression-free survival (PFS) and OS more accurately compared to the ctDNA change of an individual gene. ∆MMPM <20% was an independent predictor of PFS (2.8 vs. 14.6 months; P=0.029), although there was no significant difference for OS (16.7 vs. 17.6 months; P=0.830). In conclusion, changes in ctDNA levels were associated with anti-tumor effects, progression and disease-specific survival. ctDNA sequencing is promising for predicting response and progression after tislelizumab immunotherapy as second-line monotherapy for advanced ESCC [the present study was part of the RATIONALE-302 study (ClinicalTrials.gov identifier no. NCT03430843; 29.01.2018)].
Collapse
Affiliation(s)
- Qiong He
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Xun Shi
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Junrong Yan
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210032, P.R. China
| | - Mengmeng Wu
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210032, P.R. China
| | - Cuiping Gu
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Xinmin Yu
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
70
|
Van Baelen K, Van Cauwenberge J, Maetens M, Beck G, Camden A, Chase MC, Fraser V, Freeney S, Hutcheson L, Levine JK, Lien T, Terveer R, Turner C, Senkus E, Jankowitz RC, Vandecaveye V, Floris G, Neven P, Wildiers H, Sawyer E, Vincent-Salomon A, Derksen PWB, Desmedt C. Reporting on invasive lobular breast cancer in clinical trials: a systematic review. NPJ Breast Cancer 2024; 10:23. [PMID: 38509112 PMCID: PMC10954721 DOI: 10.1038/s41523-024-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
Invasive lobular breast cancer (ILC) differs from invasive breast cancer of no special type in many ways. Evidence on treatment efficacy for ILC is, however, lacking. We studied the degree of documentation and representation of ILC in phase III/IV clinical trials for novel breast cancer treatments. Trials were identified on Pubmed and clinicaltrials.gov. Inclusion/exclusion criteria were reviewed for requirements on histological subtype and tumor measurability. Documentation of ILC was assessed and ILC inclusion rate, central pathology and subgroup analyses were evaluated. Inclusion restrictions concerning tumor measurability were found in 39/93 manuscripts. Inclusion rates for ILC were documented in 13/93 manuscripts and varied between 2.0 and 26.0%. No central pathology for ILC was reported and 3/13 manuscripts had ILC sub-analyses. ILC is largely disregarded in most trials with poor representation and documentation. The current inclusion criteria using RECIST v1.1, fall short in recognizing the unique non-measurable metastatic infiltration of ILC.
Collapse
Affiliation(s)
- Karen Van Baelen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Josephine Van Cauwenberge
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gabriela Beck
- European Lobular Breast Cancer Consortium, Utrecht, the Netherlands
| | - Ann Camden
- Lobular Breast Cancer Alliance inc., Plymouth, MA, USA
| | | | | | - Siobhan Freeney
- European Lobular Breast Cancer Consortium, Utrecht, the Netherlands
- Lobular Ireland, Dublin, Ireland
| | | | | | - Tone Lien
- European Lobular Breast Cancer Consortium, Utrecht, the Netherlands
| | - Rian Terveer
- European Lobular Breast Cancer Consortium, Utrecht, the Netherlands
- Borstkankervereniging Nederland, Utrecht, the Netherlands
| | - Claire Turner
- European Lobular Breast Cancer Consortium, Utrecht, the Netherlands
- Lobular Breast Cancer UK, Manchester, UK
| | - Elzbieta Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Rachel C Jankowitz
- Division of Hematology/Oncology, Abramsom Cancer Center, University of Pennsylvania, Philadelphia, USA
| | - Vincent Vandecaveye
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
- Division of Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Elinor Sawyer
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Cancer Centre, King's College London, London, UK
| | - Anne Vincent-Salomon
- Department of Pathology, Institut Curie, Paris Sciences Lettres University, Paris, France
| | - Patrick W B Derksen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
71
|
Han F, Wang Y, Dong X, Lin Q, Wang Y, Gao W, Yun M, Li Y, Gao S, Huang H, Li N, Luo T, Luo X, Qiu M, Zhang D, Yan K, Li A, Liu Z. Clinical sonochemotherapy of inoperable pancreatic cancer using diagnostic ultrasound and microbubbles: a multicentre, open-label, randomised, controlled trial. Eur Radiol 2024; 34:1481-1492. [PMID: 37796294 DOI: 10.1007/s00330-023-10210-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVES Sonochemotherapy, which uses microbubble (MB)-assisted ultrasound (US) to deliver chemotherapeutic agents, has the potential to enhance tumour chemotherapy. The combination of US and MB has been demonstrated to prolong the survival of patients with pancreatic cancer. This phase 2 clinical trial aimed to determine the clinical efficacy and safety of sonochemotherapy for inoperable pancreatic ductal adenocarcinoma by using US and MB. METHODS Eighty-two patients with stage III or IV pancreatic cancer were recruited from July 2018 to March 2021 and followed up until September 2022. US treatment was performed with a modified diagnostic US scanner for 30 min after chemotherapeutic infusion. The primary endpoint was overall survival (OS), and the secondary endpoints were Eastern Cooperative Oncology Group (ECOG) status < 2, progression-free survival (PFS), disease control rate (DCR), and adverse events. RESULTS Seventy-eight patients were randomly allocated (40 to chemotherapy and 38 to sonochemotherapy). The median OS was longer with sonochemotherapy than with chemotherapy (9.10 vs. 6.10 months; p = 0.037). The median PFS with sonochemotherapy was 5.50 months, compared with 3.50 months (p = 0.080) for chemotherapy. The time of ECOG status < 2 was longer with sonochemotherapy (7.20 months) than with chemotherapy (5.00 months; p = 0.029). The DCR was 73.68% for sonochemotherapy compared with 42.50% for the control (p = 0.005). The incidence of overall adverse events was balanced between the two groups. CONCLUSIONS The use of sonochemotherapy can extend the survival and well-being time of stage III or IV pancreatic cancer patients without any increase in serious adverse events. TRIAL REGISTRATION ChineseClinicalTrials.gov ChiCTR2100044721 CLINICAL RELEVANCE STATEMENT: This multicentre, randomised, controlled trial has proven that sonochemotherapy, namely, the combination of diagnostic ultrasound, microbubbles, and chemotherapy, could extend the overall survival of patients with end-stage pancreatic ductal adenocarcinoma from 6.10 to 9.10 months without increasing any serious adverse events. KEY POINTS • This is the first multicentre, randomised, controlled trial of sonochemotherapy for clinical pancreatic cancer treatment using ultrasound and a commercial ultrasound contrast agent. • Sonochemotherapy extended the median overall survival from 6.10 (chemotherapy alone) to 9.10 months. • The disease control rate increased from 42.50% with chemotherapy to 73.68% with sonochemotherapy.
Collapse
Affiliation(s)
- Feng Han
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China
| | - Yanjie Wang
- Department of Ultrasound, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 of Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xiaoxiao Dong
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qingguang Lin
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China
| | - Yixi Wang
- Department of Ultrasound, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 of Fucheng Road, Haidian District, Beijing, 100142, China
| | - Wenhong Gao
- Department of Ultrasound, General Hospital of Central Theater, Wuhan, China
| | - Miao Yun
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China
| | - Yan Li
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shunji Gao
- Department of Ultrasound, General Hospital of Central Theater, Wuhan, China
| | - Huilong Huang
- Department of Ultrasound, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ningshan Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiao Luo
- Department of Radiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Miaozhen Qiu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dongsheng Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kun Yan
- Department of Ultrasound, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 of Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Anhua Li
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China.
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
72
|
Escobar-Mendiola D, Mediavilla-Santos L, Pérez-Mañanes R, Cuervo-Dehesa M, Vaquero-Martin J, Calvo-Haro J. Extremity high-grade sarcomas in elderly patients, are they candidates for surgery? Experience in a sarcoma referral center. Indian J Surg Oncol 2024; 15:11-18. [PMID: 38545577 PMCID: PMC10963351 DOI: 10.1007/s13193-022-01652-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/16/2022] [Indexed: 11/30/2022] Open
Abstract
The number of elderly patients diagnosed with cancer is increasing. However, knowledge regarding cancer in elderly patients is very scarce. The aim of this study is to analyze the differences in management and outcomes of high-grade extremity sarcomas between elderly and middle-aged patients. Two cohorts were made (> 70 and 30/50 years old) among patients treated in a multidisciplinary unit of a national reference center between 2011 and 2017 with a minimum of 2 years of follow-up. The management and outcomes between these two cohorts were compared. Seventy patients were included, 34 young patients and 36 elderly patients. The only difference between the treatment schemes was the chemotherapy used, 67.6% of the young patients received chemotherapy versus 16.7% of the elderly patients (p = 0.000015). There were no differences either in the overall survival or the progression-free survival between groups at 1 and 2-year follow-up. Deceases for other causes were nearly exclusive of elderly patients at a median of 45.57 months. Surgery is the treatment of choice for sarcomas in both elderly and young patients having similar results in terms of progression-free survival, overall survival, and surgery outcomes. The use of chemotherapy is the only difference in the treatment schemes between both groups. In the elderly patient with minimal or no comorbidity and good functional reserve, surgical curative treatment should be chosen. As for frail elderly patients, the therapeutic objectives must be focused mainly on quality of life and palliation of symptoms.
Collapse
Affiliation(s)
- Daniel Escobar-Mendiola
- Servicio de Cirugía Ortopédica Y Traumatología, Hospital General Universitario Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - Lydia Mediavilla-Santos
- Servicio de Cirugía Ortopédica Y Traumatología, Hospital General Universitario Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - Rubén Pérez-Mañanes
- Servicio de Cirugía Ortopédica Y Traumatología, Hospital General Universitario Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón Y Cajal, s/n, 28040 Madrid, Spain
| | - Miguel Cuervo-Dehesa
- Servicio de Cirugía Ortopédica Y Traumatología, Hospital General Universitario Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - Javier Vaquero-Martin
- Servicio de Cirugía Ortopédica Y Traumatología, Hospital General Universitario Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón Y Cajal, s/n, 28040 Madrid, Spain
| | - José Calvo-Haro
- Servicio de Cirugía Ortopédica Y Traumatología, Hospital General Universitario Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón Y Cajal, s/n, 28040 Madrid, Spain
| |
Collapse
|
73
|
Kus T, Cicin I. A perspective: the integration of ctDNA into Response Evaluation Criteria in Solid Tumours 1.1 for phase II immunotherapy clinical trials. Immunotherapy 2024; 16:319-329. [PMID: 38197142 DOI: 10.2217/imt-2023-0184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
A consensus guideline, iRECIST, was developed by the Response Evaluation Criteria in Solid Tumours (RECIST) working group for the use of the modified RECIST version 1.1 in cancer immunotherapy trials. iRECIST was designed to separate pseudoprogression from real progression. However, this is not the only ambiguous situation. In clinical immunotherapy trials, stable disease may reflect three tumor responses, including real stable disease, progressive disease and responsive disease. The prediction of a "true complete/partial response" is also important. Much data has accumulated showing that ctDNA can guide decisions at this point; thus, integrating ctDNA into the RECIST 1.1 criteria may help to distinguish a true tumor response type earlier in patients treated with immunotherapy; however, prospectively designed validation studies are needed.
Collapse
Affiliation(s)
- Tulay Kus
- School of Medicine, Department of Medical Oncology, Gaziantep University, Gaziantep, 27310, Turkey
| | - Irfan Cicin
- Department of Medical Oncology, Istinye University Topkapı Health Sciences Campus, Istanbul, 34295, Turkey
| |
Collapse
|
74
|
Batra A, Glick D, Valdes M. Contemporary Systemic Therapy Intensification for Prostate Cancer: A Review for General Practitioners in Oncology. Curr Oncol 2024; 31:1047-1062. [PMID: 38392072 PMCID: PMC10888029 DOI: 10.3390/curroncol31020078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/04/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024] Open
Abstract
Prostate cancer accounts for a significant proportion of cancer diagnoses in Canadian men. Over the past decade, the therapeutic landscape for the management of metastatic prostate cancer has undergone rapid changes. Novel strategies use hormonal agents, chemotherapy, homologous recombination repair inhibitors, and radioligand therapy or combination strategies in addition to androgen deprivation therapy. In this review, we summarize the available data addressing key therapeutic areas along the disease continuum and focus on practical aspects for general practitioners in oncology managing patients with metastatic prostate cancer.
Collapse
Affiliation(s)
- Anupam Batra
- Department of Oncology, Grand River Regional Cancer Centre, 835 King St. W., Kitchener, ON N2G 1G3, Canada; (D.G.); (M.V.)
| | | | | |
Collapse
|
75
|
Russo I, Marino D, Cozzolino C, Del Fiore P, Nerjaku F, Finotto S, Cattelan A, Calabrò ML, Belloni Fortina A, Russano F, Mazza M, Galuppo S, Bezzon E, Sbaraglia M, Krengli M, Brunello A, Mocellin S, Piaserico S, Alaibac M. Kaposi's Sarcoma: Evaluation of Clinical Features, Treatment Outcomes, and Prognosis in a Single-Center Retrospective Case Series. Cancers (Basel) 2024; 16:691. [PMID: 38398082 PMCID: PMC10887034 DOI: 10.3390/cancers16040691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Kaposi's sarcoma (KS) is a rare angioproliferative tumor classified in four different clinical-epidemiological forms. The diagnosis is based on histopathological and immunohistochemical analyses. The treatment is heterogeneous and includes several local and systemic therapeutic strategies. Methods: This is a retrospective cohort study including 86 KS patients treated between 1993 and 2022 at the University Hospital of Padua (AOPD) and at the Veneto Institute of Oncology (IOV). The data were extracted from an electronic database. Survival curves were generated using the Kaplan-Meier method, and Cox regression models were employed to explore associations with overall and disease-free survival. The male sex (89.53%), classical variant (43.02%), and cutaneous involvement (77.9%) were predominant. More than 61.6% of patients received a single treatment. Surgery, antiretroviral therapy, and chemotherapy were the mostly adopted approaches. A persistent response was observed in approximately 65% of patients, with a 22% relapse rate (at least 2 years). The overall survival ranges from 90 to 70% at 2 to 10 years after the diagnosis. Iatrogenic KS demonstrated a higher mortality (52.9%). This study reflects our experience in the management of KS. Comorbidities are very frequent, and treatments are heterogeneous. A multidisciplinary approach involving multiple referral specialists is essential for the appropriate management of this disease during diagnosis, treatment, and follow-up.
Collapse
Affiliation(s)
- Irene Russo
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (I.R.); (C.C.); (F.R.); (M.M.); (S.M.)
| | - Dario Marino
- Oncology 1 Unit, Department of Oncology, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy (S.F.)
| | - Claudia Cozzolino
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (I.R.); (C.C.); (F.R.); (M.M.); (S.M.)
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (I.R.); (C.C.); (F.R.); (M.M.); (S.M.)
| | - Fitnete Nerjaku
- Department of Medicine (DIMED), School of Medicine, University of Padova, 35128 Padova, Italy; (F.N.); (M.S.)
| | - Silvia Finotto
- Oncology 1 Unit, Department of Oncology, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy (S.F.)
| | - Annamaria Cattelan
- Infectious and Tropical Diseases Unit, Padova University Hospital, 35128 Padova, Italy;
| | - Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy;
| | - Anna Belloni Fortina
- Dermatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (A.B.F.); (S.P.); (M.A.)
- Pediatric Dermatology Regional Center, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy
| | - Francesco Russano
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (I.R.); (C.C.); (F.R.); (M.M.); (S.M.)
| | - Marcodomenico Mazza
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (I.R.); (C.C.); (F.R.); (M.M.); (S.M.)
| | - Sara Galuppo
- Radiotherapy Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (S.G.); (M.K.)
| | - Elisabetta Bezzon
- Radiology Unit, Veneto Institute of Oncology, IOV – IRCCS, 35128 Padova, Italy
| | - Marta Sbaraglia
- Department of Medicine (DIMED), School of Medicine, University of Padova, 35128 Padova, Italy; (F.N.); (M.S.)
- Department of Pathology, Azienda Ospedale—University of Padova, 35128 Padova, Italy
| | - Marco Krengli
- Radiotherapy Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (S.G.); (M.K.)
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128 Padova, Italy
| | - Antonella Brunello
- Oncology 1 Unit, Department of Oncology, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy (S.F.)
| | - Simone Mocellin
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV – IRCCS, 35128 Padova, Italy; (I.R.); (C.C.); (F.R.); (M.M.); (S.M.)
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128 Padova, Italy
| | - Stefano Piaserico
- Dermatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (A.B.F.); (S.P.); (M.A.)
| | - Mauro Alaibac
- Dermatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (A.B.F.); (S.P.); (M.A.)
| |
Collapse
|
76
|
Salaffi F, Mazzei MA, Aliprandi A, Martino F, Moretti B, Silvestri E, Di Meglio N, Bagnacci G, Di Carlo M, Sinigaglia L, Gerli R, Tranquilli Leali P, Faletti C, Giovagnoni A. Structured request form in musculoskeletal radiology examinations (CONCERTO): results of an expert Delphi consensus-structured radiology request form for correct classification of patients to undergo radiological examinations of the Italian Society of Medical and Interventional Radiology (SIRM), the Italian Society of Rheumatology (SIR) and the Italian Society of Orthopedics and Traumatology (SIOT). LA RADIOLOGIA MEDICA 2024; 129:307-314. [PMID: 38315280 PMCID: PMC10879249 DOI: 10.1007/s11547-024-01762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024]
Abstract
PURPOSE To describe a Delphi consensus for the realization of a structured radiology request form for patients undergoing musculoskeletal imaging. METHODS A steering committee (four radiologists, a rheumatologist and an orthopedic surgeon) proposed a form to an expert panel (30 members, ten radiologists, ten rheumatologists and ten orthopedic surgeons). Through an online survey, the panelists voted on their level of agreement with the statements of the form using a 10-point Likert scale (1: no agreement; 10: total agreement) in a three-round process. A combination of two distinct criteria, a mean agreement level ≥ 8 and a percentage of at least 75% of responses with a value ≥ 8, was deemed as acceptable. RESULTS The form achieved high median ratings in all the assessed key features. During the first round, all items met the threshold to be advanced as unmodified in the next round. Additional proposed items were considered and introduced in the next round (six items in Section 1, five items in Section 2, ten items in Section 3, 11 items in Section 4, six items in Section 5, eight items in Section 6, ten items in Section 7 and eight items in Section 8). Of these items, in round 3, only six reached the threshold to be integrated into the final form. CONCLUSIONS Implementation of a structured radiology request form can improve appropriateness and collaboration between clinicians and radiologists in musculoskeletal imaging.
Collapse
Affiliation(s)
- Fausto Salaffi
- Rheumatology Clinic, Università Politecnica delle Marche, "Carlo Urbani" Hospital, Via Aldo Moro, 25, 60035, Jesi, Ancona, Italy
| | - Maria Antonietta Mazzei
- Unit of Diagnostic Imaging, Department of Medical, Surgical and Neurosciences, Department of Radiological Sciences, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alberto Aliprandi
- Unit of Radiology, Clinical Institutes Zucchi, Via Bartolomeo Zucchi, 24, 20052, Monza, Italy
| | | | - Biagio Moretti
- Department of Translational Biomedicine and Neuroscience ("DibraiN")-Operative unit of Orthopaedics and Traumatology, University General Hospital, Bari, Italy
| | - Enzo Silvestri
- Diagnostica per Immagini, Istituto Salus-Alliance Medical, Genoa, Italy
| | - Nunzia Di Meglio
- Unit of Diagnostic Imaging, Department of Medical, Surgical and Neurosciences, Department of Radiological Sciences, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Giulio Bagnacci
- Unit of Diagnostic Imaging, Department of Medical, Surgical and Neurosciences, Department of Radiological Sciences, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Marco Di Carlo
- Rheumatology Clinic, Università Politecnica delle Marche, "Carlo Urbani" Hospital, Via Aldo Moro, 25, 60035, Jesi, Ancona, Italy.
| | - Luigi Sinigaglia
- Specialista in Reumatologia e Medicina Interna, Casa di Cura "La Madonnina", Milan, Italy
| | - Roberto Gerli
- Department of Medicine, Rheumatology Unit, University of Perugia, Perugia, Italy
| | | | - Carlo Faletti
- , Dipartimento per Immagini dell'A.O. C.T.O-C.R.F.- M, Turin, Italy
| | - Andrea Giovagnoni
- Department of Radiology, University Hospital "Azienda Ospedaliera Universitaria delle Marche", Ancona, Italy
| |
Collapse
|
77
|
Zhang J, Wang Z, Zhu L, Wang C, Huang B, Zhong Y, Ying P, Wang H, Li Q, Feng L, Wang X, Jin H. Estrogen receptor α inhibits Caveolin 1 translation by promoting m6A-dependent miR199a-5p maturation to confer nab-paclitaxel resistance. Am J Cancer Res 2023; 13:6210-6225. [PMID: 38187046 PMCID: PMC10767350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 01/09/2024] Open
Abstract
Estrogen receptor positive (ER+) breast cancer patients exhibit poorer responsiveness to nab-paclitaxel compared to ER negative (ER-) patients, with the underlying mechanisms remaining unknown. Caveolin 1 (CAV1) is a membrane invagination protein critical for the endocytosis of macromolecules including albumin-bound chemotherapeutic agents. Here, we demonstrate that ERα limits the efficacy of nab-paclitaxel in breast cancer cells while genetic or pharmacological inhibition of ERα increased the sensitivity of ER+ breast cancer cells to nab-paclitaxel. Notably, CAV1 expression inversely correlates with ERα and relates to improved clinical outcomes from nab-paclitaxel treatment. Importantly, ERα stimulates m6A dependent maturation of miR199a-5p, which is elevated in ER+ breast cancer, to inhibit CAV1 translation by antagonizing m6A modification of CAV1 mRNA. Together, our findings reveal a novel role of ERα in promoting m6A modification and subsequent maturation of miR199a-5p, which is upregulated in ER+ breast cancer, leading to the suppression of m6A modification of CAV1 and its mRNA translation, thereby contributing to nab-paclitaxel resistance. Thus, combining an ER antagonist with nab-paclitaxel could offer a promising strategy for treating ER+ breast cancer patients.
Collapse
Affiliation(s)
- Jianping Zhang
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Zhuo Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Liyuan Zhu
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Chaoqun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical UniversityDongyang, Zhejiang, China
| | - Bifei Huang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical UniversityDongyang, Zhejiang, China
| | - Yiming Zhong
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Pingting Ying
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Hanying Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Qinglin Li
- The Cancer Hospital of The University of Chinese Academy of SciencesHangzhou, Zhejiang, China
| | - Lifeng Feng
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Hongchuan Jin
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| |
Collapse
|
78
|
Kim J, Lee Y. Mobocertinib and Bevacizumab for Amivantamab-Refractory Lung Cancer With EGFR Exon 20 Insertion Mutation: A Case Report. JTO Clin Res Rep 2023; 4:100600. [PMID: 38124788 PMCID: PMC10730360 DOI: 10.1016/j.jtocrr.2023.100600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/20/2023] [Accepted: 11/01/2023] [Indexed: 12/23/2023] Open
Abstract
Amivantamab is the first drug approved in EGFR exon 20 insertion-mutated NSCLC. Nevertheless, primary or secondary resistance to amivantamab is a frequent problem in clinical practice. We report a case of a patient with EGFR exon 20-mutated NSCLC who had primary resistance to amivantamab but was successfully treated by combining therapy of another EGFR exon 20 insertion-specific targeted drug mobocertinib and bevacizumab.
Collapse
Affiliation(s)
- Jaemin Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Youngjoo Lee
- Research Institute, National Cancer Center, Goyang, Republic of Korea
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
79
|
Honoré N, van der Elst A, Dietz A, van Marcke C, Helaers R, Mendola A, Dahou H, Marbaix E, Poncin R, Seront E, Schmitz S, Limaye N, Galot R, Machiels JP. Tumour-agnostic plasma assay for circulating tumour DNA predicts outcome in recurrent and/or metastatic squamous cell carcinoma of the head and neck treated with a PD-1 inhibitor. Eur J Cancer 2023; 195:113372. [PMID: 37913682 DOI: 10.1016/j.ejca.2023.113372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Only 15-20% of recurrent and/or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN) patients derive long-term benefit from nivolumab or pembrolizumab. We developed a circulating tumour DNA (ctDNA) tumour-agnostic assay aimed at the early prediction of single agent programmed cell death 1 (PD1) inhibitor efficacy in R/M SCCHN. PATIENTS AND METHODS Our tumour-agnostic assay included 37 genes frequently mutated in R/M SCCHN and two HPV16 genes. Primary endpoint was the concordance between ctDNA kinetics (ΔctDNA) and the best overall response according to Response Evaluation Criteria in Solid Tumors version 1.1. ΔctDNA was defined as the difference in mean variant allele frequency (VAF) between the on-treatment sample harvested 6-10 weeks (FU1) after PD1 inhibitor initiation and the pre-treatment plasma sample (ΔctDNA = mean FU1 VAF - mean pre-treatment VAF). RESULTS ctDNA was detected in 35/44 (80%) of the pre-treatment plasma samples. The concordance between ΔctDNA and imaging response was observed in 74%. Median progression-free survival was 8.6 months in the favourable ΔctDNA group and 2.5 months in the unfavourable ΔctDNA group (p = 0.057). Median overall survival (OS) was 18.1 and 8.2 months in the favourable and unfavourable ΔctDNA groups, respectively (p = 0.13). In patients with PD-L1 expressing SCCHN (Combined Positive Score ≥1), OS was significantly better in patients with favourable ΔctDNA compared with patients with unfavourable ΔctDNA: median OS was 41.5 and 8.4 months (p = 0.033), respectively. CONCLUSIONS Tumour-agnostic ctDNA analysis for human papillomavirus (HPV)-negative and HPV-positive R/M SCCHN is feasible. ctDNA kinetics show promising results in predicting the efficacy of PD1 inhibitors in R/M SCCHN.
Collapse
Affiliation(s)
- Natasha Honoré
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Athenaïs van der Elst
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Anna Dietz
- Department of Radiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Cédric van Marcke
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Raphael Helaers
- Human Molecular Genetics, de Duve Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Antonella Mendola
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Hajar Dahou
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Etienne Marbaix
- Department of Pathology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Renaud Poncin
- Department of Medical Oncology, Cliniques Saint-Pierre, Ottignies, Belgium
| | - Emanuel Seront
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Medical Oncology, Jolimont Hospital, La Louviere, Belgium
| | - Sandra Schmitz
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of ENT and Maxillo-facial Surgery, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Nisha Limaye
- Genetics of Autoimmune Diseases and Cancer, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Rachel Galot
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Pascal Machiels
- Pôle Oncologie, Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
80
|
Zeng N, Wang Y, Wan Y, Wang H, Li N. The Antitumor Impact of Combining Hepatic Artery Ligation With Copper Chelators for Liver Cancer. Clin Med Insights Oncol 2023; 17:11795549231204612. [PMID: 38023286 PMCID: PMC10666691 DOI: 10.1177/11795549231204612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/13/2023] [Indexed: 12/01/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the main cancer-related mortality worldwide. Thus, there is a constant search for improvement in treatment strategies to enhance the prognosis of this malignancy. The study aims to investigate the combined antitumor activity of ammonium tetrathiomolybdate (TM, copper chelator) combined with hepatic artery ligation (HAL) for liver cancer. Methods A total of 40 Sprague-Dawley (SD) rats bearing hepatic tumors were randomly divided into four groups: the control group without any treatment (control), HAL only (HAL), given TM by gavage (TM), and given TM combined with HAL (HAL + TM). The concentrations of serum copper were measured at the predetermined time points. Tumor growth rate, overall survival (OS), expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and microvessel density (MVD), as determined by immunohistochemical examination, were compared. Results HAL treatment transiently could elevate alanine transaminase (ALT) and aspartate transaminase (AST) but resumed to baseline within 1 week. Serum copper was significantly increased in tumor-bearing animals over time. The values of serum copper in the three treatment groups were significantly lower than those in the control group at different time points, with the lowest values observed in the TM group (P < .05). The average tumor size was 30.33 ± 2.58, 20.83 ± 2.93, 16.80 ± 3.84, and 10.88 ± 1.08 mm in the control, HAL, TM, and HAL + TM groups, respectively (HAL + TM vs other groups, all P < .05). In addition, the expression levels of HIF-1α, VEGF, and MVD were significantly lower in the HAL + TM group than those in the other groups (P < .05). The OS of rats in the combined groups was significantly prolonged combined to the other groups (P < .05), with survival time of 19.1 ± 0.64, 25.4 ± 1.24, 25.3 ± 1.78, and 29.9 ± 2.22 days in the control, HAL, TM, and HAL + TM groups, respectively. Conclusion These findings suggest that combined treatment with TM and HAL holds great potential for liver cancer treatment by reducing tumor hypoxia and angiogenesis. The observed results indicate that these combinations may offer a novel target and strategy for interventional therapy of liver cancer.
Collapse
Affiliation(s)
- Ni Zeng
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ye Wang
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuan Wan
- Interventional Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hongyu Wang
- Department of Interventional Therapy, Guangdong Provincial Hospital of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Nan Li
- Department of Interventional Radiology, Guangzhou First People’s Hospital, Guangzhou, China
| |
Collapse
|
81
|
Aggarwal R, Starzinski S, de Kouchkovsky I, Koshkin V, Bose R, Chou J, Desai A, Kwon D, Kaushal S, Trihy L, Rastogi M, Ippisch R, Aslam M, Friedlander T, Feng F, Oh D, Cheung A, Small E, Evans M, Fong L, Hope TA. Single-dose 177Lu-PSMA-617 followed by maintenance pembrolizumab in patients with metastatic castration-resistant prostate cancer: an open-label, dose-expansion, phase 1 trial. Lancet Oncol 2023; 24:1266-1276. [PMID: 37922930 PMCID: PMC10667020 DOI: 10.1016/s1470-2045(23)00451-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/20/2023] [Accepted: 09/06/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Checkpoint inhibitors have been shown to have limited activity in patients with metastatic castration-resistant prostate cancer. We aimed to determine whether a single dose of lutetium-177 [177Lu]-prostate-specific membrane antigen (PSMA)-617 (177Lu-PSMA-617) followed by maintenance pembrolizumab was safe and could induce durable clinical benefit. METHODS We did an open-label, dose-expansion, phase 1 study at the University of California, San Francisco (San Fransisco, CA, USA). Eligible patients were men aged 18 years or older with progressive metastatic castration-resistant prostate cancer who had an Eastern Cooperative Oncology Group performance status of 0 or 1, had progression on one or more androgen signalling inhibitors, and at least three PSMA-avid lesions on 68Ga-PSMA-11 positron emission tomography. In part A, patients were enrolled sequentially to one of three schedules in which a single dose of 177Lu-PSMA-617 (7·4 GBq) was given intravenously 28 days before (schedule 1), concomitant with (schedule 2), or 21 days after (schedule 3) the start of maintenance intravenous pembrolizumab (200 mg every 3 weeks). In part B, 25 patients were enrolled using the recommended phase 2 schedule. The primary endpoint in part A was determination of the recommended phase 2 schedule, and in part B, the objective response rate. The analysis set included all patients who received at least one dose of pembrolizumab or 177Lu-PSMA-617. This study is registered with ClinicalTrials.gov, NCT03805594. FINDINGS Between Aug 8, 2019 and May 7, 2022, 43 male patients were enrolled (n=18 part A [six patients per schedule]; n=25 part B), with a median follow-up of 16·5 months (IQR 12·2-21·9). Schedule 1 was selected as the recommended phase 2 schedule for part B, on the basis of safety and feasibility of administration observed in part A. In part B, 14 (56%; 95% CI 35-76) of 25 patients had a confirmed objective response. Two (5%) of 43 patients had a treatment-related adverse event of grade 3 or worse (grade 3 arthritis in schedule 2, grade 3 pneumonitis in schedule 3). One serious adverse event (one death due to aspiration pneumonia) and no treatment-related deaths were observed. INTERPRETATION A single priming dose of 177Lu-PSMA-617 followed by pembrolizumab maintenance was safe and had encouraging preliminary activity in patients with metastatic castration-resistant prostate cancer. FUNDING Prostate Cancer Foundation, National Cancer Institute, Novartis Pharmaceuticals, and Merck.
Collapse
Affiliation(s)
- Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| | - Stephanie Starzinski
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Ivan de Kouchkovsky
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Vadim Koshkin
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Rohit Bose
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan Chou
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Arpita Desai
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Daniel Kwon
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Samuel Kaushal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Lauren Trihy
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Medini Rastogi
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Robin Ippisch
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Maya Aslam
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Terence Friedlander
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Felix Feng
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - David Oh
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Alexander Cheung
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Eric Small
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Michael Evans
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
82
|
Donners R, Candito A, Blackledge M, Rata M, Messiou C, Koh DM, Tunariu N. Repeatability of quantitative individual lesion and total disease multiparametric whole-body MRI measurements in prostate cancer bone metastases. Br J Radiol 2023; 96:20230378. [PMID: 37660399 PMCID: PMC10607420 DOI: 10.1259/bjr.20230378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/07/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
OBJECTIVES To assess the repeatability of quantitative multiparametric whole-body MRI (mpWB-MRI) parameters in advanced prostate cancer (APC) bone metastases. METHODS 1.5T MRI was performed twice on the same day in 10 APC patients. MpWB-MRI-included diffusion weighted imaging (DWI) and T1-weighted gradient-echo 2-point Dixon sequences. ADC and relative fat-fraction percentage (rFF%) maps were calculated, respectively. A radiologist delineated up to 10 target bone metastases per study. Means of ADC, b900 signal intensity(SI), normalised b900 SI, rFF% and maximum diameter (MD) for each target lesion and overall parameter averages across all targets per patient were recorded. The total disease volume (tDV in ml) was manually delineated on b900 images and mean global (g)ADC was derived. Bland-Altman analyses were performed with calculation of 95% repeatability coefficients (RC). RESULTS Seventy-three individual targets (median MD 26 mm) were included. Lesion mean ADC RC was 12.5%, mean b900 SI RC 137%, normalised mean b900 SI RC 110%, rFF% RC 3.2 and target MD RC 5.5 mm (16.3%). Patient target lesion average mean ADC RC was 6.4%, b900 SI RC 104% and normalised mean b900 SI RC 39.6%. Target average rFF% RC was 1.8, average MD RC 1.3 mm (4.8%). tDV segmentation RC was 6.4% and mean gADC RC 5.3%. CONCLUSIONS APC bone metastases' ADC, rFF% and maximum diameter, tDV and gADC show good repeatability. ADVANCES IN KNOWLEDGE APC bone metastases' mean ADC and rFF% measurements of single lesions and global disease volumes are repeatable, supporting their potential role as quantitative biomarkers in metastatic bone disease.
Collapse
Affiliation(s)
| | - Antonio Candito
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, Sutton, United Kingdom
| | - Matthew Blackledge
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, Sutton, United Kingdom
| | - Mihaela Rata
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, Sutton, United Kingdom
| | | | | | | |
Collapse
|
83
|
Tomonari T, Tani J, Sato Y, Tanaka H, Tanaka T, Taniguchi T, Kawano Y, Morishita A, Okamoto K, Sogabe M, Miyamoto H, Masaki T, Takayama T. Clinical Features and Outcomes of Conversion Therapy in Patients with Unresectable Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:5221. [PMID: 37958395 PMCID: PMC10650115 DOI: 10.3390/cancers15215221] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
This retrospective multicenter study analyzed 244 patients with unresectable hepatocellular carcinoma treated with lenvatinib (LEN) and atezolizumab + bevacizumab (Atezo + Bev) to examine the characteristics, treatment courses, and prognoses. The cases of patients who could achieve HCC downstaging from Barcelona Clinic Liver Cancer (BCLC) stage B or C to A or zero indicated the need for conversion therapy. The patients' prognoses with and without conversion therapy were compared. Of the 244 patients, 12 (4.9%) underwent conversion therapy, six out of 131 (4.6%) were treated with LEN, and six out of 113 (5.3%) were treated with Atezo + Bev. Eleven patients (91.7%) with a modified albumin bilirubin (mALBI) grade 1 or 2a and BCLC-B stage showed significantly higher rates of transition during conversion therapy (p < 0.05). The patients undergoing conversion therapy had a significantly longer median overall survival rate than those receiving chemotherapy alone (1208 [1064-NA] vs. 569 [466-704] days, p < 0.01). A comparison of the patients who achieved a partial response with and without conversion was evaluated using propensity score matching to reduce the confounding factors, showing a significant survival benefit in the conversion group (1208 [1064-NA] vs. 665 days, p < 0.01). Among the patients with u-HCC who were treated with LEN and Atezo + Bev, those with mALBI 1 + 2a and BCLC-B were likely to achieve conversion therapy with downstaging.
Collapse
Affiliation(s)
- Tetsu Tomonari
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa University Graduate School of Medicine, Kagawa 761-0701, Japan; (J.T.); (A.M.); (T.M.)
| | - Yasushi Sato
- Department of Community Medicine for Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8503, Japan
| | - Hironori Tanaka
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Takahiro Tanaka
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Tatsuya Taniguchi
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University Graduate School of Medicine, Kagawa 761-0701, Japan; (J.T.); (A.M.); (T.M.)
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University Graduate School of Medicine, Kagawa 761-0701, Japan; (J.T.); (A.M.); (T.M.)
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima 770-8504, Japan; (T.T.); (H.T.); (T.T.); (T.T.); (Y.K.); (K.O.); (M.S.); (H.M.); (T.T.)
| |
Collapse
|
84
|
He LN, Chen T, Fu S, Jiang Y, Zhang X, Chen C, Du W, Luo L, Li A, Wang Y, Yu H, Zhou Y, Wang Y, Yang Y, Huang Y, Zhao H, Fang W, Zhang L, Hong S. Tumor response assessment by measuring the single largest lesion per organ in advanced non-small cell lung cancer patients treated with PD-1/PD-L1 inhibitor. Ther Adv Med Oncol 2023; 15:17588359231200463. [PMID: 37881238 PMCID: PMC10594961 DOI: 10.1177/17588359231200463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 08/25/2023] [Indexed: 10/27/2023] Open
Abstract
Background For Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST1.1), measuring up to two target lesions per organ is an arbitrary criterion. Objectives We sought to compare response assessment using RECIST1.1 and modified RECIST1.1 (mRECIST1.1, measuring the single largest lesion per organ) in advanced non-small cell lung cancer (aNSCLC) patients undergoing anti-PD-1/PD-L1 monotherapy. Methods Concordance of radiologic response categorization between RECIST1.1 and mRECIST1.1 was compared using the Kappa statistics. C-index was calculated to evaluate prognostic accuracy of radiologic response by the two criteria. The Kaplan-Meier method and Cox regression analysis were conducted for progression-free survival (PFS) and overall survival (OS). Results Eighty-seven patients who had at least two target lesions in any organ per the RECIST1.1 were eligible for comparison analysis. Tumor response showed excellent concordance when measured using the RECIST1.1 and mRECIST1.1 (Kappa = 0.961). C-index by these two criteria was similar for PFS (0.784 versus 0.785) and OS (0.649 versus 0.652). Responders had significantly longer PFS and OS versus non-responders (p < 0.05), whichever criterion adopted. Radiologic response remained a significant predictor of PFS and OS in multivariate analysis (p < 0.05). Conclusion The mRECIST1.1 was comparable to RECIST1.1 in response assessment among aNSCLC patients who received single-agent PD-1/PD-L1 inhibitor. The mRECIST1.1, with reduced number of lesions to be measured, may be sufficient and more convenient to assess antitumor activity in clinical practice.
Collapse
Affiliation(s)
- Li-Na He
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tao Chen
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sha Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen University; Department of Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongluo Jiang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xuanye Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chen Chen
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Du
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Linfeng Luo
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Anlin Li
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yixing Wang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Yu
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yixin Zhou
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of VIP region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuhong Wang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong 510060, China
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong 510060, China
- Department of Oncology, The People’s Hospital of Fengqing, No. 107, Qingyun Group 1, Pingcun Village Committee, Fengshan Town, Fengqing County, Lincang 675900, China
| |
Collapse
|
85
|
Gondry O, Xavier C, Raes L, Heemskerk J, Devoogdt N, Everaert H, Breckpot K, Lecocq Q, Decoster L, Fontaine C, Schallier D, Aspeslagh S, Vaneycken I, Raes G, Van Ginderachter JA, Lahoutte T, Caveliers V, Keyaerts M. Phase I Study of [ 68Ga]Ga-Anti-CD206-sdAb for PET/CT Assessment of Protumorigenic Macrophage Presence in Solid Tumors (MMR Phase I). J Nucl Med 2023; 64:1378-1384. [PMID: 37474271 PMCID: PMC10478821 DOI: 10.2967/jnumed.122.264853] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/05/2023] [Indexed: 07/22/2023] Open
Abstract
Macrophages play an important role throughout the body. Antiinflammatory macrophages expressing the macrophage mannose receptor (MMR, CD206) are involved in disease development, ranging from oncology to atherosclerosis and rheumatoid arthritis. [68Ga]Ga-NOTA-anti-CD206 single-domain antibody (sdAb) is a PET tracer targeting CD206. This first-in-human study, as its primary objective, evaluated the safety, biodistribution, and dosimetry of this tracer. The secondary objective was to assess its tumor uptake. Methods: Seven patients with a solid tumor of at least 10 mm, an Eastern Cooperative Oncology Group score of 0 or 1, and good renal and hepatic function were included. Safety was evaluated using clinical examination and blood sampling before and after injection. For biodistribution and dosimetry, PET/CT was performed at 11, 90, and 150 min after injection; organs showing tracer uptake were delineated, and dosimetry was evaluated. Blood samples were obtained at selected time points for blood clearance. Metabolites in blood and urine were assessed. Results: Seven patients were injected with, on average, 191 MBq of [68Ga]Ga-NOTA-anti-CD206-sdAb. Only 1 transient adverse event of mild severity was considered to be possibly, although unlikely, related to the study drug (headache, Common Terminology Criteria for Adverse Events grade 1). The blood clearance was fast, with less than 20% of the injected activity remaining after 80 min. There was uptake in the liver, kidneys, spleen, adrenals, and red bone marrow. The average effective dose from the radiopharmaceutical was 4.2 mSv for males and 5.2 mSv for females. No metabolites were detected. Preliminary data of tumor uptake in cancer lesions showed higher uptake in the 3 patients who subsequently progressed than in the 3 patients without progression. One patient could not be evaluated because of technical failure. Conclusion: [68Ga]Ga-NOTA-anti-CD206-sdAb is safe and well tolerated. It shows rapid blood clearance and renal excretion, enabling high contrast-to-noise imaging at 90 min after injection. The radiation dose is comparable to that of routinely used PET tracers. These findings and the preliminary results in cancer patients warrant further investigation of this tracer in phase II clinical trials.
Collapse
Affiliation(s)
- Odrade Gondry
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Catarina Xavier
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurens Raes
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Johannes Heemskerk
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Nick Devoogdt
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hendrik Everaert
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lore Decoster
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christel Fontaine
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Denis Schallier
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sandrine Aspeslagh
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ilse Vaneycken
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Geert Raes
- Cellular and Molecular Immunology, Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology, Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Tony Lahoutte
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Vicky Caveliers
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
86
|
Zeng Q, Xiong F, Liu L, Zhong L, Cai F, Zeng X. Radiomics Based on DCE-MRI for Predicting Response to Neoadjuvant Therapy in Breast Cancer. Acad Radiol 2023; 30 Suppl 2:S38-S49. [PMID: 37169624 DOI: 10.1016/j.acra.2023.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
RATIONALE AND OBJECTIVES To compare the value of radiomics and diameter% based on pre- and early-treatment dynamic enhanced MR (DCE-MRI) of the breast in predicting response to neoadjuvant therapy (NAT) in breast cancer and to construct a tool for early noninvasive prediction of NAT outcomes. MATERIALS AND METHODS Retrospective analysis of clinical and imaging data of 142 patients with primary invasive breast cancer who underwent DCE-MRI before and after two cycles of NAT at our institution. Enroled patients were randomly assigned in a 7:3 ratio to the training group and the test group. Patients were divided into pathological complete response (pCR) and non-pathological complete response groups based on surgical pathology findings after NAT. The maximum diameter relative regression values (Diameter%) before and after treatment were calculated and the conventional imaging Diameter% model was constructed. Based on pre- and early-NAT DCE-MRI, the optimal features of pre-NAT, early-NAT, and delta radiomics were screened using redundancy analysis, least absolute shrinkage, and selection operator methods to construct the corresponding radiomics model and calculate the Radscores. Indicators that were statistically significant in the univariate analysis of clinical data were further screened by stepwise regression and combined with Radscores to construct the fusion model. All models were evaluated and compared. RESULTS In the test set, the area under the curve (AUC) of the delta radiomics model (0.87) was higher than that of the pre-NAT, early-NAT radiomics models (0.57, 0.78) and the Diameter% model (0.83). The fusion model had the best efficacy in predicting pCR after NAT, with AUCs of 0.91 in the training and test sets. And its nomogram plot showed that Radscore of early-NAT radiomics had the greatest weight. In the test set, the fusion model and Delta radiomics model improved the efficacy of predicting pCR by 35.56% and 14.19%, respectively, compared to the Diameter% model (P = 0 and .039). Clinical decision curves showed the highest overall clinical benefit for the fusion model. CONCLUSION Radiomics, especially delta and early-NAT radiomics, may be potential biomarkers for early noninvasive prediction of NAT outcomes. And a fusion model constructed from meaningful clinicopathological indicators combined with radiomics can effectively predict NAT response.
Collapse
Affiliation(s)
- Qiao Zeng
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Q.Z., F.C., X.Z.); Department of Radiology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China (Q.Z., L.L., L.Z.); Department of Radiology, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China (Q.Z., L.L., L.Z.)
| | - Fei Xiong
- Department of Ultrasound, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang, China (F.X.)
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China (Q.Z., L.L., L.Z.); Department of Radiology, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China (Q.Z., L.L., L.Z.)
| | - Linhua Zhong
- Department of Radiology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China (Q.Z., L.L., L.Z.); Department of Radiology, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China (Q.Z., L.L., L.Z.)
| | - Fengqin Cai
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Q.Z., F.C., X.Z.)
| | - Xianjun Zeng
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Q.Z., F.C., X.Z.).
| |
Collapse
|
87
|
Zhu C, Li H, Yang X, Wang S, Wang Y, Zhang N, Wang Y, Xue J, Zhang L, Ning C, Yang X, Xun Z, Chao J, Long J, Sang X, Zhu Z, Zhao H. Efficacy, safety, and prognostic factors of PD-1 inhibitors combined with lenvatinib and Gemox chemotherapy as first-line treatment in advanced intrahepatic cholangiocarcinoma: a multicenter real-world study. Cancer Immunol Immunother 2023; 72:2949-2960. [PMID: 37247023 PMCID: PMC10412480 DOI: 10.1007/s00262-023-03466-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/10/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND A programmed cell death protein-1 (PD-1) inhibitor combined with lenvatinib and Gemox chemotherapy as first-line therapy demonstrated high anti-tumor activity against biliary tract cancer in phase II clinical trials. Herein, we aimed to investigate the efficacy and safety for advanced intrahepatic cholangiocarcinoma (ICC) in a multicenter real-world study. METHODS Patients with advanced ICC who received PD-1 inhibitor combined with lenvatinib and Gemox chemotherapy were retrospectively screened at two medical centers. The primary endpoints were overall survival (OS) and progression-free survival (PFS), whereas the secondary endpoints were objective response rate (ORR), disease control rate (DCR), and safety. Prognostic factors for survival were analyzed. RESULTS Fifty-three patients with advanced ICC were included in this study. The median follow-up time was 13.7 (95% confidence interval (CI): 12.9-17.2) months. The median OS and PFS were 14.3 (95% CI: 11.3-NR) and 8.63 (95% CI: 7.17-11.6) months, respectively. The ORR, DCR, and clinical benefit rate were 52.8, 94.3, and 75.5%, respectively. In the multivariate analysis, the tumor burden score (TBS), tumor-node metastasis classification (TNM) stage, and PD-L1 expression were independent prognostic factors for OS and PFS. All patients experienced adverse events (AEs), 41.5% (22/53) experienced grade 3 or 4 AEs, including fatigue (8/53, 15.1%) and myelosuppression (7/53, 13.2%). No grade 5 AEs were reported. CONCLUSION PD-1 inhibitors combined with lenvatinib and Gemox chemotherapy represent an effective and tolerable regimen for advanced ICC in a multicenter retrospective real-world study. TBS, TNM stage, and PD-L1 expression can be used as potential prognostic factors for OS and PFS.
Collapse
Affiliation(s)
- Chengpei Zhu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Hu Li
- Department of Hepatobiliary Surgery, The Fifth Medical Center of PLA General Hospital, No. 100 Middle Road of West 4th Ring Road, Fengtai District, Beijing, 100039, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yunchao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yanyu Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jingnan Xue
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Cong Ning
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xu Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Ziyu Xun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jiashuo Chao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xinting Sang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Zhenyu Zhu
- Department of Hepatobiliary Surgery, The Fifth Medical Center of PLA General Hospital, No. 100 Middle Road of West 4th Ring Road, Fengtai District, Beijing, 100039, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| |
Collapse
|
88
|
Zhang J, Guo Y, Zhou J, Rasmussen HE. PubPredict: Prediction of progression and survival in oncology leveraging publications and early efficacy data. Pharm Stat 2023; 22:963-973. [PMID: 37439295 DOI: 10.1002/pst.2321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 05/15/2023] [Accepted: 06/20/2023] [Indexed: 07/14/2023]
Abstract
In oncology/hematology early phase clinical trials, efficacies were often observed in terms of response rate, depth, timing, and duration. However, the true clinical benefits that eventually support registrational purpose are progression-free survival (PFS) and/or overall survival (OS), the follow-up of which are typically not long enough in early phase trials. This gap imposes challenges in strategies for late phase drug development. In this article, we tackle the question by leveraging published study to establish a quantitative link between early efficacy outcomes and late phase efficacy endpoints. We used solid tumor cancer as disease model. We modeled the disease course of a RECISTv1.1 assessed solid tumor with a continuous Markov chain (CMC) model. We parameterize the transition intensity matrix of a CMC model based on published aggregate-level summary statistics, and then simulate subject-level time-to-event data. The simulated data is shown to have good approximation to published studies. PFS and/or OS could be predicted with the transition intensity matrix modified given clinical knowledge to reflect various assumptions on response rate, depth, timing, and duration. The authors have built a R shiny application named PubPredict, the tool implements the algorithm described above and allows customized features including multiple response levels, treatment crossover and varying follow-up duration. This toolset has been applied to advise phase 3 trial design when only early efficacy data are available from phase 1 or 2 studies.
Collapse
Affiliation(s)
| | - Yufei Guo
- Department of Statistics, George Washington University, Washington, DC, USA
| | - Junyi Zhou
- Amgen Inc., Thousand Oaks, California, USA
| | | |
Collapse
|
89
|
Tanaka K, Tsuji K, Hiraoka A, Tada T, Hirooka M, Kariyama K, Tani J, Atsukawa M, Takaguchi K, Itobayashi E, Fukunishi S, Ishikawa T, Tajiri K, Ochi H, Toyoda H, Ogawa C, Nishimura T, Hatanaka T, Kakizaki S, Shimada N, Kawata K, Naganuma A, Kosaka H, Matono T, Kuroda H, Yata Y, Ohama H, Tada F, Nouso K, Morishita A, Tsutsui A, Nagano T, Itokawa N, Okubo T, Arai T, Yokohama K, Nishikawa H, Imai M, Koizumi Y, Nakamura S, Iijima H, Kaibori M, Hiasa Y, Kumada T. Usefulness of Tumor Marker Score for Predicting the Prognosis of Hepatocellular Carcinoma Patients Treated with Atezolizumab Plus Bevacizumab: A Multicenter Retrospective Study. Cancers (Basel) 2023; 15:4348. [PMID: 37686624 PMCID: PMC10486534 DOI: 10.3390/cancers15174348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
AIM This study aimed to evaluate the ability of a previously reported tumor marker (TM) score involving alpha-fetoprotein (AFP), fucosylated AFP (AFP-L3), and des gamma-carboxy prothrombin (DCP) as TMs in predicting the prognosis and therapeutic efficacy in hepatocellular carcinoma (HCC) patients administered atezolizumab plus bevacizumab (Atez/Bev) as first-line treatment. MATERIALS/METHODS The study period covered September 2020 to December 2022 and involved 371 HCC patients treated with Atez/Bev. The values of the TMs AFP, AFP-L3, and DCP were measured upon introducing Atez/Bev. Elevations in the values of AFP (≥100 ng/mL), AFP-L3 (≥10%), and DCP (≥100 mAU/mL) were considered to indicate a positive TM. The number of positive TMs was summed up and used as the TM score, as previously proposed. Hepatic reserve function was assessed using the modified albumin-bilirubin grade (mALBI). Predictive values for prognosis were evaluated retrospectively. RESULTS A TM score of 0 was shown in 81 HCC patients (21.8%), 1 in 110 (29.6%), 2 in 112 (29.9%), and 3 in 68 (18.3%). The median overall survival (OS) times for TM scores 0, 1, 2, and 3 were not applicable [NA] (95% CI NA-NA), 24.0 months (95% CI 17.8-NA), 16.7 months (95% CI 17.8-NA), and NA (95% CI 8.3-NA), respectively (p < 0.001). The median progression-free survival (PFS) times for TM scores 0, 1, 2, and 3 were 16.5 months (95% CI 8.0-not applicable [NA]), 13.8 months (95% CI 10.6-21.3), 7.7 months (95% CI 5.3-8.9), and 5.8 months (95% CI 3.0-7.6), respectively (p < 0.001). OS was well stratified in mALBI 1/2a and mALBI 2a/2b. PFS was well stratified in mALBI 2a/2b, but not in mALBI 1/2a. CONCLUSIONS The TM score involving AFP, AFP-L3, and DCP as TMs was useful in predicting the prognosis and therapeutic efficacy in terms of OS and PFS in HCC patients administered Atez/Bev as first-line treatment.
Collapse
Affiliation(s)
- Kazunari Tanaka
- Center for Gastroenterology, Teine Keijinkai Hospital, Sapporo 006-8555, Japan;
| | - Kunihiko Tsuji
- Center for Gastroenterology, Teine Keijinkai Hospital, Sapporo 006-8555, Japan;
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama 790-0024, Japan; (H.O.); (F.T.)
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji 670-8540, Japan; (T.T.); (S.N.)
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (M.H.); (Y.K.); (Y.H.)
| | - Kazuya Kariyama
- Department of Hepatology, Okayama City Hospital, Okayama 700-0962, Japan; (K.K.); (K.N.)
| | - Joji Tani
- Department of Gastroenterology and Hepatology, Kagawa University, Kagawa 761-0795, Japan; (J.T.); (A.M.)
| | - Masanori Atsukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo 113-8603, Japan (N.I.); (T.O.); (T.A.)
| | - Koichi Takaguchi
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu 760-8557, Japan; (K.T.); (A.T.); (T.N.)
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Asahi 289-2511, Japan;
| | - Shinya Fukunishi
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (S.F.); (K.Y.); (H.N.)
| | - Toru Ishikawa
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata 950-1104, Japan; (T.I.); (M.I.)
| | - Kazuto Tajiri
- Department of Gastroenterology, Toyama University Hospital, Toyama 930-0194, Japan;
| | - Hironori Ochi
- Center for Liver-Biliary-Pancreatic Disease, Matsuyama Red Cross Hospital, Matsuyama 790-8524, Japan;
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki 503-8502, Japan;
| | - Chikara Ogawa
- Department of Gastroenterology, Japanese Red Cross Takamatsu Hospital, Takamatsu 760-0017, Japan;
| | - Takashi Nishimura
- Department of Gastroenterology and Hepatology, Hyogo Medical University, Nishinomiya 663-8501, Japan; (T.N.); (H.I.)
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi 371-0821, Japan;
| | - Satoru Kakizaki
- Department of Clinical Research, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan;
| | - Noritomo Shimada
- Division of Gastroenterology and Hepatology, Otakanomori Hospital, Kashiwa 277-0863, Japan;
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu 565-0871, Japan;
| | - Atsushi Naganuma
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan;
| | - Hisashi Kosaka
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.K.); (M.K.)
| | - Tomomitsu Matono
- Department of Hepatology, St. Mary’s Hospital, Himeji 670-0801, Japan;
| | - Hidekatsu Kuroda
- Division of Hepatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka 020-8505, Japan;
| | - Yutaka Yata
- Department of Gastroenterology, Hanwa Memorial Hospital, Osaka 558-0041, Japan;
| | - Hideko Ohama
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama 790-0024, Japan; (H.O.); (F.T.)
| | - Fujimasa Tada
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama 790-0024, Japan; (H.O.); (F.T.)
| | - Kazuhiro Nouso
- Department of Hepatology, Okayama City Hospital, Okayama 700-0962, Japan; (K.K.); (K.N.)
| | - Asahiro Morishita
- Department of Gastroenterology and Hepatology, Kagawa University, Kagawa 761-0795, Japan; (J.T.); (A.M.)
| | - Akemi Tsutsui
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu 760-8557, Japan; (K.T.); (A.T.); (T.N.)
| | - Takuya Nagano
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu 760-8557, Japan; (K.T.); (A.T.); (T.N.)
| | - Norio Itokawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo 113-8603, Japan (N.I.); (T.O.); (T.A.)
| | - Tomomi Okubo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo 113-8603, Japan (N.I.); (T.O.); (T.A.)
| | - Taeang Arai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo 113-8603, Japan (N.I.); (T.O.); (T.A.)
| | - Keisuke Yokohama
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (S.F.); (K.Y.); (H.N.)
| | - Hiroki Nishikawa
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (S.F.); (K.Y.); (H.N.)
| | - Michitaka Imai
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata 950-1104, Japan; (T.I.); (M.I.)
| | - Yohei Koizumi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (M.H.); (Y.K.); (Y.H.)
| | - Shinichiro Nakamura
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji 670-8540, Japan; (T.T.); (S.N.)
| | - Hiroko Iijima
- Department of Gastroenterology and Hepatology, Hyogo Medical University, Nishinomiya 663-8501, Japan; (T.N.); (H.I.)
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan; (H.K.); (M.K.)
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (M.H.); (Y.K.); (Y.H.)
| | - Takashi Kumada
- Department of Nursing, Gifu Kyoritsu University, Ogaki 503-8550, Japan;
| |
Collapse
|
90
|
Ju H, Wei D, Wu Y, Liu Y, Ding Q, Rui M, Fan Z, Yao Y, Hu J, Ren G. A pilot study of camrelizumab with docetaxel and cisplatin for the first line treatment in recurrent/metastatic oral squamous cell carcinoma. MedComm (Beijing) 2023; 4:e312. [PMID: 37492783 PMCID: PMC10363852 DOI: 10.1002/mco2.312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 07/27/2023] Open
Abstract
Pembrolizumab with cisplatin and 5-fluorouracil showed survival benefit but relatively high occurrence of treatment-related adverse events (TRAEs) for recurrent/metastatic oral squamous cell carcinoma (R/M OSCC). A more tolerable regime is needed. This trial enrolled 20 R/M OSCC patients with previously untreated and PD-L1 positive. Patients were administered camrelizumab with docetaxel and cisplatin every 3 weeks for six cycles, followed by camrelizumab monotherapy every 3 weeks until disease progression or intolerable toxicity. The primary endpoint was occurrence of grade ≥ 3 TRAEs, secondary endpoints included overall survival (OS), progression-free survival (PFS), and overall response rate (ORR). 45% patients experienced grade ≥ 3 TRAEs, which the most common were anemia (15%), stomatitis (15%), and neutropenia (10%). The most common potential immune-related adverse events were reactive cutaneous capillary endothelial proliferation (RCCEP; 60%), hypothyroidism (35%), and pneumonitis (15%). No treatment-related deaths occurred. The median OS, PFS, and ORR was 14.4 months, 5.35 months, and 40.0% respectively. The study also found RCCEP occurrence, lower FOXP3+ cells, and higher density of intratumor tertiary lymphoid structure were associated with improved efficacy. Our data suggest that camrelizumab with docetaxel/cisplatin as first-line therapy was well tolerable and had potentially favorite efficacy in PD-L1-positive patients with R/M OSCC.
Collapse
Affiliation(s)
- Houyu Ju
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Dongliang Wei
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Yunteng Wu
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Yang Liu
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Qi Ding
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- School of StomatologyWeifang Medical UniversityWeifangChina
| | - Mengyu Rui
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Zongyu Fan
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Yanli Yao
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Jingzhou Hu
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| | - Guoxin Ren
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiChina
- National Clinical Research Center of StomatologyShanghaiChina
| |
Collapse
|
91
|
Ando Y, Kawaoka T, Kosaka M, Shirane Y, Johira Y, Miura R, Murakami S, Yano S, Amioka K, Naruto K, Kosaka Y, Uchikawa S, Kodama K, Fujino H, Nakahara T, Ono A, Murakami E, Yamauchi M, Okamoto W, Takahashi S, Imamura M, Aikata H. Risk Factors for Early Onset of Proteinuria in Patients Receiving Atezolizumab Plus Bevacizumab for Unresectable Hepatocellular Carcinoma. Liver Cancer 2023; 12:251-261. [PMID: 37601981 PMCID: PMC10433089 DOI: 10.1159/000528145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/22/2022] [Indexed: 08/22/2023] Open
Abstract
Introduction Proteinuria is one of the adverse events of atezolizumab plus bevacizumab combination therapy (Atezo + Bev) and can cause interruption in the use of Bev. However, the risk factors for proteinuria in patients with hepatocellular carcinoma (HCC) who are receiving Atezo + Bev have not yet been investigated. The aim of this study was to identify the risk factors for early onset of proteinuria in Atezo + Bev for patients with unresectable HCC. Methods Sixty-four patients with Child-Pugh scores of 5-7, an Eastern Cooperative Oncology Group performance status of 0 or 1, and low level of proteinuria (1+ or less on a dipstick test and urine protein-to-creatinine ratio (UPCR) less than 2.0 g/g Cr) at the initiation of therapy were analyzed. The level of proteinuria was evaluated based on the Common Terminology Criteria for Adverse Events version 5.0. We adopted the UPCR for the quantitative test instead of a 24-h urine collection. The incidence of proteinuria and changes in liver function were retrospectively investigated. Results The cumulative incidence of proteinuria over a 24-week period was 34.4%. Multivariate analysis showed that a low estimated glomerular filtration rate (hazard ratio [HR], 3.807; 95% confidence interval [CI], 1.579-9.180; p = 0.003), treatment for hypertension (HR, 6.224; 95% CI, 1.614-24.010; p = 0.008), and high systolic blood pressure (SBP) (HR, 2.649; 95% CI, 1.133-6.194; p = 0.025) were risk factors for proteinuria. Serum albumin levels and albumin-bilirubin scores in patients with proteinuria worsened. In addition, a mean SBP ≥135 mm Hg during treatment was the only risk factor for the development of severe proteinuria (UPCR >2 g/g Cr). Conclusion Our study found that controlling blood pressure is extremely important for the management of proteinuria in patients with HCC who are receiving Atezo + Bev.
Collapse
Affiliation(s)
- Yuwa Ando
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masanari Kosaka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Shirane
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yusuke Johira
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ryoichi Miura
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Serami Murakami
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeki Yano
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kei Amioka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kensuke Naruto
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yumi Kosaka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinsuke Uchikawa
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kenichiro Kodama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hatsue Fujino
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Nakahara
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atushi Ono
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eisuke Murakami
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masami Yamauchi
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Clinical Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Wataru Okamoto
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Clinical Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shoichi Takahashi
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
92
|
Cao L, Chen S, Sun R, Ashby CR, Wei L, Huang Z, Chen ZS. Darovasertib, a novel treatment for metastatic uveal melanoma. Front Pharmacol 2023; 14:1232787. [PMID: 37576814 PMCID: PMC10419210 DOI: 10.3389/fphar.2023.1232787] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
The FDA granted orphan drug designation to darovasertib, a first-in-class oral, small molecular inhibitor of protein kinase C (PKC), for the treatment of uveal melanoma, on 2 May 2022. Primary uveal melanoma has a high risk of progressing to metastatic uveal melanoma, with a poor prognosis. The activation of the PKC and mitogen-activated protein kinase pathways play an essential role in the pathogenesis of uveal melanoma, and mutations in the G protein subunit alpha q (GNAQ), and G protein subunit alpha11 (GNA11) genes are considered early events in the development of uveal melanoma. Compared to other PKC inhibitors, such as sotrastaurin and enzastaurin, darovasertib is significantly more potent in inhibiting conventional (α, β) and novel (δ, ϵ, η, θ) PKC proteins and has a better tolerability and safety profile. Current Phase I/II clinical trials indicated that darovasertib, combined with the Mitogen-activated protein kinase/Extracellular (MEK) inhibitors, binimetinib or crizotinib, produced a synergistic effect of uveal melanoma. In this article, we summarize the development of drugs for treating uveal melanomas and discuss problems associated with current treatments. We also discuss the mechanism of action, pharmacokinetic profile, adverse effects, and clinical trial for darovasertib, and future research directions for treating uveal melanoma.
Collapse
Affiliation(s)
- Lei Cao
- Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Shuzhen Chen
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Rainie Sun
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
- Stuyvesant High School, New York, NY, United States
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| |
Collapse
|
93
|
Gouel P, Callonnec F, Levêque É, Valet C, Blôt A, Cuvelier C, Saï S, Saunier L, Pepin LF, Hapdey S, Libraire J, Vera P, Viard B. Evaluation of the capability and reproducibility of RECIST 1.1. measurements by technologists in breast cancer follow-up: a pilot study. Sci Rep 2023; 13:9148. [PMID: 37277412 DOI: 10.1038/s41598-023-36315-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
The evaluation of tumor follow-up according to RECIST 1.1 has become essential in clinical practice given its role in therapeutic decision making. At the same time, radiologists are facing an increase in activity while facing a shortage. Radiographic technologists could contribute to the follow-up of these measures, but no studies have evaluated their ability to perform them. Ninety breast cancer patients were performed three CT follow-ups between September 2017 and August 2021. 270 follow-up treatment CT scans were analyzed including 445 target lesions. The rate of agreement of classifications RECIST 1.1 between five technologists and radiologists yielded moderate (k value between 0.47 and 0.52) and substantial (k value = 0.62 and k = 0.67) agreement values. 112 CT were classified as progressive disease (PD) by the radiologists, and 414 new lesions were identified. The analysis showed a percentage of strict agreement of progressive disease classification between reader-technologists and radiologists ranging from substantial to almost perfect agreement (range 73-97%). Analysis of intra-observer agreement was strong at almost perfect (k > 0.78) for 3 technologists. These results are encouraging regarding the ability of selected technologists to perform measurements according to RECIST 1.1 criteria by CT scan with good identification of disease progression.
Collapse
Affiliation(s)
- Pierrick Gouel
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France.
- QuantIF-LITIS EA4108, University of Rouen, Rouen, Normandy, France.
| | - Françoise Callonnec
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Émilie Levêque
- Department of Statistics and Clinical Research Unit, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Céline Valet
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Axelle Blôt
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Clémence Cuvelier
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Sonia Saï
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Lucie Saunier
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Louis-Ferdinand Pepin
- Department of Statistics and Clinical Research Unit, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Sébastien Hapdey
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
- QuantIF-LITIS EA4108, University of Rouen, Rouen, Normandy, France
| | - Julie Libraire
- Department of Statistics and Clinical Research Unit, Henri Becquerel Cancer Center, Rouen, Normandy, France
| | - Pierre Vera
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
- QuantIF-LITIS EA4108, University of Rouen, Rouen, Normandy, France
| | - Benjamin Viard
- Department of Medical Imaging, Henri Becquerel Cancer Center, Rouen, Normandy, France
| |
Collapse
|
94
|
Kepenek F, Kömek H, Can C, Kaplan İ, Altindağ S, Gündoğan C. The prognostic role of whole-body volumetric 68 GA-DOTATATE PET/computed tomography parameters in patients with gastroenteropancreatic neuroendocrine tumor treated with 177 LU-DOTATATE. Nucl Med Commun 2023; 44:509-517. [PMID: 37038931 DOI: 10.1097/mnm.0000000000001693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
OBJECTIVE The aim of this study is to evaluate the prognostic role of Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 and quantitative 68 Ga-DOTATATE PET/computed tomography parameters such as maximum standardized uptake value (SUVmax), mean SUV (SUVmean), DOTATATE tumor volume (DTV), total lesion DOTATATE (TLD) in patients with gastroenteropancreatic neuroendocrine tumors (GEP-NETs) treated with 177 Lu-DOTATATE. MATERIAL AND METHOD Our retrospective study included 21 patients with GEP-NETs treated with 177 Lu-DOTATATE between January 2017 and January 2022. SUVmax, SUVmean, SUVmax/spleenSUVmax (SUVmax/Sx), DTV, TLD, SUVmean/spleenSUVmean (SUVmean/Sm), TLD/Sm values were calculated and recorded for all patients before and after 177 Lu-DOTATATE treatment. RESULTS A total of 319 metastases were detected in the patients included in the study, and a total of 68 target lesions were selected. In univariant Cox regression analysis, TLD/Sm percent change (∆TLD/Sm) was found to be statistically significant on overall survival (OS) ( P = 0.044). The 3-year survival in nonresponders was 50% ( P = 0.034) based on ∆SUVmax/Sx values, 50% ( P = 0.002) based on RECIST values, 50% based on ∆TDTV + new lesion values ( P = 0.033), and according to ∆TTLD + new lesion values, it was 66% ( P = 0.030). CONCLUSION In our study, we showed that SUVmax/Sx, RECIST, ∆TDTV + new lesion, and ∆TTLD + new lesion parameters can predict OS in the evaluation of response to treatment.
Collapse
Affiliation(s)
- Ferat Kepenek
- Department of of Nuclear Medicine, University of Health Sciences, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakir
| | - Halil Kömek
- Department of of Nuclear Medicine, University of Health Sciences, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakir
| | - Canan Can
- Department of of Nuclear Medicine, University of Health Sciences, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakir
| | - İhsan Kaplan
- Department of of Nuclear Medicine, University of Health Sciences, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakir
| | - Serdar Altindağ
- Department of Nuclear Medicine, İstanbul Gelisim University Vocational College of Health Services Hisar Intercontinental Hospital, İstanbul, Turkey
| | - Cihan Gündoğan
- Department of of Nuclear Medicine, University of Health Sciences, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakir
| |
Collapse
|
95
|
Jost-Brinkmann F, Demir M, Wree A, Luedde T, Loosen SH, Müller T, Tacke F, Roderburg C, Mohr R. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma: Results from a German real-world cohort. Aliment Pharmacol Ther 2023; 57:1313-1325. [PMID: 36883351 DOI: 10.1111/apt.17441] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 02/18/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND AND AIMS Phase III trials have established atezolizumab plus bevacizumab as the novel standard of care for patients with unresectable hepatocellular carcinoma (HCC). However, these trials raised concerns regarding treatment efficacy in non-viral HCC, and it remains unclear whether combination immunotherapy is safe and effective in patients with advanced cirrhosis. METHODS One hundred patients with unresectable HCC initiated therapy with atezolizumab plus bevacizumab at our centre between January 2020 and March 2022. The control cohort consisted of 80 patients with advanced HCC who received either sorafenib (n = 43) or lenvatinib (n = 37) as systemic treatment. RESULTS Overall survival (OS) and progression-free survival (PFS) were significantly longer within the atezolizumab/bevacizumab group and comparable to phase III data. The benefits in terms of increased objective response rate (ORR), OS and PFS were consistent across subgroups, including non-viral HCC (58%). The ROC-optimised neutrophil-to-lymphocyte ratio (NLR) cut-off of 3.20 was the strongest independent predictor of ORR and PFS. In patients with advanced cirrhosis Child-Pugh B, liver function was significantly better preserved with immunotherapy. Patients with Child-Pugh B cirrhosis showed similar ORR but shorter OS and PFS compared to patients with preserved liver function. CONCLUSIONS Atezolizumab plus bevacizumab showed good efficacy and safety in patients with unresectable HCC and partially advanced liver cirrhosis in a real-world setting. Moreover, the NLR was able to predict response to atezolizumab/bevacizumab treatment and may guide patient selection.
Collapse
Affiliation(s)
- Fabian Jost-Brinkmann
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Alexander Wree
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Sven H Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tobias Müller
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| |
Collapse
|
96
|
Lother D, Robert M, Elwood E, Smith S, Tunariu N, Johnston SRD, Parton M, Bhaludin B, Millard T, Downey K, Sharma B. Imaging in metastatic breast cancer, CT, PET/CT, MRI, WB-DWI, CCA: review and new perspectives. Cancer Imaging 2023; 23:53. [PMID: 37254225 DOI: 10.1186/s40644-023-00557-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Breast cancer is the most frequent cancer in women and remains the second leading cause of death in Western countries. It represents a heterogeneous group of diseases with diverse tumoral behaviour, treatment responsiveness and prognosis. While major progress in diagnosis and treatment has resulted in a decline in breast cancer-related mortality, some patients will relapse and prognosis in this cohort of patients remains poor. Treatment is determined according to tumor subtype; primarily hormone receptor status and HER2 expression. Menopausal status and site of disease relapse are also important considerations in treatment protocols. MAIN BODY Staging and repeated evaluation of patients with metastatic breast cancer are central to the accurate assessment of disease extent at diagnosis and during treatment; guiding ongoing clinical management. Advances have been made in the diagnostic and therapeutic fields, particularly with new targeted therapies. In parallel, oncological imaging has evolved exponentially with the development of functional and anatomical imaging techniques. Consistent, reproducible and validated methods of assessing response to therapy is critical in effectively managing patients with metastatic breast cancer. CONCLUSION Major progress has been made in oncological imaging over the last few decades. Accurate disease assessment at diagnosis and during treatment is important in the management of metastatic breast cancer. CT (and BS if appropriate) is generally widely available, relatively cheap and sufficient in many cases. However, several additional imaging modalities are emerging and can be used as adjuncts, particularly in pregnancy or other diagnostically challenging cases. Nevertheless, no single imaging technique is without limitation. The authors have evaluated the vast array of imaging techniques - individual, combined parametric and multimodal - that are available or that are emerging in the management of metastatic breast cancer. This includes WB DW-MRI, CCA, novel PET breast cancer-epitope specific radiotracers and radiogenomics.
Collapse
Affiliation(s)
| | - Marie Robert
- Institut de Cancérologie de l'Ouest, St Herblain, France
| | | | - Sam Smith
- The Royal Marsden Hospital, London & Sutton, UK
| | - Nina Tunariu
- The Royal Marsden Hospital, London & Sutton, UK
- The Institute of Cancer Research (ICR), London & Sutton, UK
| | - Stephen R D Johnston
- The Royal Marsden Hospital, London & Sutton, UK
- The Institute of Cancer Research (ICR), London & Sutton, UK
| | | | | | | | - Kate Downey
- The Royal Marsden Hospital, London & Sutton, UK
- The Institute of Cancer Research (ICR), London & Sutton, UK
| | - Bhupinder Sharma
- The Royal Marsden Hospital, London & Sutton, UK.
- The Institute of Cancer Research (ICR), London & Sutton, UK.
| |
Collapse
|
97
|
Granata V, Fusco R, Villanacci A, Grassi F, Grassi R, Di Stefano F, Petrone A, Fusco N, Ianniello S. Qualitative and semi-quantitative ultrasound assessment in delta and Omicron Covid-19 patients: data from high volume reference center. Infect Agent Cancer 2023; 18:34. [PMID: 37245026 DOI: 10.1186/s13027-023-00515-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
OBJECTIVE to evaluate the efficacy of US, both qualitatively and semi-quantitatively, in the selection of treatment for the Covid-19 patient, using patient triage as the gold standard. METHODS Patients admitted to the Covid-19 clinic to be treated with monoclonal antibodies (mAb) or retroviral treatment and undergoing lung ultrasound (US) were selected from the radiological data set between December 2021 and May 2022 according to the following inclusion criteria: patients with proven Omicron variant and Delta Covid-19 infection; patients with known Covid-19 vaccination with at least two doses. Lung US (LUS) was performed by experienced radiologists. The presence, location, and distribution of abnormalities, such as B-lines, thickening or ruptures of the pleural line, consolidations, and air bronchograms, were evaluated. The anomalous findings in each scan were classified according to the LUS scoring system. Nonparametric statistical tests were performed. RESULTS The LUS score median value in the patients with Omicron variant was 1.5 (1-20) while the LUS score median value in the patients with Delta variant was 7 (3-24). A difference statistically significant was observed for LUS score values among the patients with Delta variant between the two US examinations (p value = 0.045 at Kruskal Wallis test). There was a difference in median LUS score values between hospitalized and non-hospitalized patients for both the Omicron and Delta groups (p value = 0.02 on the Kruskal Wallis test). For Delta patients groups the sensitivity, specificity, positive and negative predictive values, considering a value of 14 for LUS score for the hospitalization, were of 85.29%, 44.44%, 85.29% and 76.74% respectively. CONCLUSIONS LUS is an interesting diagnostic tool in the context of Covid-19, it could allow to identify the typical pattern of diffuse interstitial pulmonary syndrome and could guide the correct management of patients.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", 80131, Naples, Italy
| | | | - Alberta Villanacci
- Department of Radiology and Diagnostic Imaging, National Institute for Infectious Diseases IRCCS Lazzaro Spallanzani, 00149, Rome, Italy
| | - Francesca Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122, Milan, Italy
| | - Roberta Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122, Milan, Italy
| | - Federica Di Stefano
- Department of Radiology and Diagnostic Imaging, National Institute for Infectious Diseases IRCCS Lazzaro Spallanzani, 00149, Rome, Italy
| | - Ada Petrone
- Department of Radiology and Diagnostic Imaging, National Institute for Infectious Diseases IRCCS Lazzaro Spallanzani, 00149, Rome, Italy
| | - Nicoletta Fusco
- Department of Radiology and Diagnostic Imaging, National Institute for Infectious Diseases IRCCS Lazzaro Spallanzani, 00149, Rome, Italy
| | - Stefania Ianniello
- Department of Radiology and Diagnostic Imaging, National Institute for Infectious Diseases IRCCS Lazzaro Spallanzani, 00149, Rome, Italy
| |
Collapse
|
98
|
Tanabe N, Saeki I, Aibe Y, Matsuda T, Hanazono T, Nishi M, Hidaka I, Kuwashiro S, Shiratsuki S, Matsuura K, Egusa M, Nishiyama N, Fujioka T, Kawamoto D, Sasaki R, Nishimura T, Oono T, Hisanaga T, Matsumoto T, Ishikawa T, Yamasaki T, Takami T. Early Prediction of Response Focused on Tumor Markers in Atezolizumab plus Bevacizumab Therapy for Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:2927. [PMID: 37296889 PMCID: PMC10251947 DOI: 10.3390/cancers15112927] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Despite the promising efficacy of atezolizumab plus bevacizumab (atezo/bev), some patients with unresectable hepatocellular carcinoma (HCC) experience disease progression. This retrospective study, which included 154 patients, aimed to evaluate predictors of treatment efficacy of atezo/bev for unresectable HCC. Factors associated with treatment response were examined, focusing on tumor markers. In the high-alpha-fetoprotein (AFP) group (baseline AFP ≥ 20 ng/mL), a decrease in AFP level > 30% was an independent predictor of objective response (odds ratio, 5.517; p = 0.0032). In the low-AFP group (baseline AFP < 20 ng/mL), baseline des-gamma-carboxy prothrombin (DCP) level < 40 mAU/mL was an independent predictor of objective response (odds ratio, 3.978; p = 0.0206). The independent predictors of early progressive disease were an increase in AFP level ≥ 30% at 3 weeks (odds ratio, 4.077; p = 0.0264) and the presence of extrahepatic spread (odds ratio, 3.682; p = 0.0337) in the high-AFP group and up-to-seven criteria, OUT (odds ratio, 15.756; p = 0.0257) in the low-AFP group. In atezo/bev therapy, focusing on early AFP changes, baseline DCP, and tumor burden of up-to-seven criteria are useful in predicting response to treatment.
Collapse
Affiliation(s)
- Norikazu Tanabe
- Division of Laboratory, Yamaguchi University Hospital, Ube 755-8505, Yamaguchi, Japan; (N.T.); (T.Y.)
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Issei Saeki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Yuki Aibe
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Kokura Memorial Hospital, Kitakyushu 802-8555, Fukuoka, Japan
| | - Takashi Matsuda
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Shimonoseki Medical Center, Shimonoseki 750-0061, Yamaguchi, Japan
| | - Tadasuke Hanazono
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Saiseikai Shimonoseki General Hospital, Shimonoseki 759-6603, Yamaguchi, Japan
| | - Maiko Nishi
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Yamaguchi Rosai Hospital, Sanyo-Onoda 756-0095, Yamaguchi, Japan
| | - Isao Hidaka
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Saiseikai Yamaguchi General Hospital, Yamaguchi 753-0078, Yamaguchi, Japan
| | - Shinya Kuwashiro
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Yamaguchi Prefectural Grand Medical Center, Hofu 747-8511, Yamaguchi, Japan
| | - Shogo Shiratsuki
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Tokuyama Central Hospital, Syunan 745-8522, Yamaguchi, Japan
| | - Keiji Matsuura
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Gastroenterology, Shuto General Hospital, Yanai 742-0032, Yamaguchi, Japan
| | - Maho Egusa
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Natsuko Nishiyama
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Tsuyoshi Fujioka
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Daiki Kawamoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Ryo Sasaki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Tatsuro Nishimura
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Takashi Oono
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Takuro Hisanaga
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Toshihiko Matsumoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Tsuyoshi Ishikawa
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| | - Takahiro Yamasaki
- Division of Laboratory, Yamaguchi University Hospital, Ube 755-8505, Yamaguchi, Japan; (N.T.); (T.Y.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
- Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (M.E.); (N.N.); (T.F.); (D.K.); (R.S.); (T.N.); (T.O.); (T.H.); (T.M.); (T.I.); (T.T.)
- Yamaguchi Clinical Research Group—Hepatology (YCR-H), Ube 755-8505, Yamaguchi, Japan (M.N.); (I.H.); (S.K.)
| |
Collapse
|
99
|
Laqua FC, Woznicki P, Bley TA, Schöneck M, Rinneburger M, Weisthoff M, Schmidt M, Persigehl T, Iuga AI, Baeßler B. Transfer-Learning Deep Radiomics and Hand-Crafted Radiomics for Classifying Lymph Nodes from Contrast-Enhanced Computed Tomography in Lung Cancer. Cancers (Basel) 2023; 15:2850. [PMID: 37345187 PMCID: PMC10216416 DOI: 10.3390/cancers15102850] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/06/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023] Open
Abstract
OBJECTIVES Positron emission tomography (PET) is currently considered the non-invasive reference standard for lymph node (N-)staging in lung cancer. However, not all patients can undergo this diagnostic procedure due to high costs, limited availability, and additional radiation exposure. The purpose of this study was to predict the PET result from traditional contrast-enhanced computed tomography (CT) and to test different feature extraction strategies. METHODS In this study, 100 lung cancer patients underwent a contrast-enhanced 18F-fluorodeoxyglucose (FDG) PET/CT scan between August 2012 and December 2019. We trained machine learning models to predict FDG uptake in the subsequent PET scan. Model inputs were composed of (i) traditional "hand-crafted" radiomics features from the segmented lymph nodes, (ii) deep features derived from a pretrained EfficientNet-CNN, and (iii) a hybrid approach combining (i) and (ii). RESULTS In total, 2734 lymph nodes [555 (20.3%) PET-positive] from 100 patients [49% female; mean age 65, SD: 14] with lung cancer (60% adenocarcinoma, 21% plate epithelial carcinoma, 8% small-cell lung cancer) were included in this study. The area under the receiver operating characteristic curve (AUC) ranged from 0.79 to 0.87, and the scaled Brier score (SBS) ranged from 16 to 36%. The random forest model (iii) yielded the best results [AUC 0.871 (0.865-0.878), SBS 35.8 (34.2-37.2)] and had significantly higher model performance than both approaches alone (AUC: p < 0.001, z = 8.8 and z = 22.4; SBS: p < 0.001, z = 11.4 and z = 26.6, against (i) and (ii), respectively). CONCLUSION Both traditional radiomics features and transfer-learning deep radiomics features provide relevant and complementary information for non-invasive N-staging in lung cancer.
Collapse
Affiliation(s)
- Fabian Christopher Laqua
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany
| | - Piotr Woznicki
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany
| | - Thorsten A. Bley
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany
| | - Mirjam Schöneck
- Institute of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Miriam Rinneburger
- Institute of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Mathilda Weisthoff
- Institute of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Matthias Schmidt
- Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Thorsten Persigehl
- Institute of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Andra-Iza Iuga
- Institute of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Bettina Baeßler
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
100
|
Park LK, Lim KH, Volkman J, Abdiannia M, Johnston H, Nigogosyan Z, Siegel MJ, McGill JB, McKee AM, Salam M, Zhang RM, Ma D, Popuri K, Chow VTY, Beg MF, Hawkins WG, Peterson LR, Ippolito JE. Safety, tolerability, and effectiveness of the sodium-glucose cotransporter 2 inhibitor (SGLT2i) dapagliflozin in combination with standard chemotherapy for patients with advanced, inoperable pancreatic adenocarcinoma: a phase 1b observational study. Cancer Metab 2023; 11:6. [PMID: 37202813 DOI: 10.1186/s40170-023-00306-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy. Thus, there is an urgent need for safe and effective novel therapies. PDAC's excessive reliance on glucose metabolism for its metabolic needs provides a target for metabolic therapy. Preclinical PDAC models have demonstrated that targeting the sodium-glucose co-transporter-2 (SGLT2) with dapagliflozin may be a novel strategy. Whether dapagliflozin is safe and efficacious in humans with PDAC is unclear. METHODS We performed a phase 1b observational study (ClinicalTrials.gov ID NCT04542291; registered 09/09/2020) to test the safety and tolerability of dapagliflozin (5 mg p.o./day × 2 weeks escalated to 10 mg p.o./day × 6 weeks) added to standard Gemcitabine and nab-Paclitaxel (GnP) chemotherapy in patients with locally advanced and/or metastatic PDAC. Markers of efficacy including Response Evaluation Criteria in Solid Tumors (RECIST 1.1) response, CT-based volumetric body composition measurements, and plasma chemistries for measuring metabolism and tumor burden were also analyzed. RESULTS Of 23 patients who were screened, 15 enrolled. One expired (due to complications from underlying disease), 2 dropped out (did not tolerate GnP chemotherapy) during the first 4 weeks, and 12 completed. There were no unexpected or serious adverse events with dapagliflozin. One patient was told to discontinue dapagliflozin after 6 weeks due to elevated ketones, although there were no clinical signs of ketoacidosis. Dapagliflozin compliance was 99.4%. Plasma glucagon increased significantly. Although abdominal muscle and fat volumes decreased; increased muscle-to-fat ratio correlated with better therapeutic response. After 8 weeks of treatment in the study, partial response (PR) to therapy was seen in 2 patients, stable disease (SD) in 9 patients, and progressive disease (PD) in 1 patient. After dapagliflozin discontinuation (and chemotherapy continuation), an additional 7 patients developed the progressive disease in the subsequent scans measured by increased lesion size as well as the development of new lesions. Quantitative imaging assessment was supported by plasma CA19-9 tumor marker measurements. CONCLUSIONS Dapagliflozin is well-tolerated and was associated with high compliance in patients with advanced, inoperable PDAC. Overall favorable changes in tumor response and plasma biomarkers suggest it may have efficacy against PDAC, warranting further investigation.
Collapse
Affiliation(s)
- Lauren K Park
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA
| | - Kian-Huat Lim
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jonas Volkman
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mina Abdiannia
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Hannah Johnston
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA
| | - Zack Nigogosyan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA
| | - Marilyn J Siegel
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA
| | - Janet B McGill
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Alexis M McKee
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Maamoun Salam
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Rong M Zhang
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Da Ma
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - William G Hawkins
- Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Linda R Peterson
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA.
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA.
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, Mail Stop Code: 8131, 4559 Scott Ave., St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| |
Collapse
|