201
|
A multidisciplinary consensus on the morphological and functional responses to immunotherapy treatment. Clin Transl Oncol 2020; 23:434-449. [PMID: 32623581 PMCID: PMC7936941 DOI: 10.1007/s12094-020-02442-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 06/22/2020] [Indexed: 02/08/2023]
Abstract
The implementation of immunotherapy has radically changed the treatment of oncological patients. Currently, immunotherapy is indicated in the treatment of patients with head and neck tumors, melanoma, lung cancer, bladder tumors, colon cancer, cervical cancer, breast cancer, Merkel cell carcinoma, liver cancer, leukemia and lymphomas. However, its efficacy is restricted to a limited number of cases. The challenge is, therefore, to identify which subset of patients would benefit from immunotherapy. To this end, the establishment of immunotherapy response criteria and predictive and prognostic biomarkers is of paramount interest. In this report, a group of experts of the Spanish Society of Medical Oncology (SEOM), the Spanish Society of Medical Radiology (SERAM), and Spanish Society of Nuclear Medicine and Molecular Imaging (SEMNIM) provide an up-to-date review and a consensus guide on these issues.
Collapse
|
202
|
Wang H, Mao X. Evaluation of the Efficacy of Neoadjuvant Chemotherapy for Breast Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2423-2433. [PMID: 32606609 PMCID: PMC7308147 DOI: 10.2147/dddt.s253961] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
Neoadjuvant chemotherapy is increasingly used in breast cancer, especially for downstaging the primary tumor in the breast and the metastatic axillary lymph node. Accurate evaluations of the response to neoadjuvant chemotherapy provide important information on the impact of systemic therapies on breast cancer biology, prognosis, and guidance for further therapy. Moreover, pathologic complete response is a validated and valuable surrogate prognostic factor of survival after therapy. Evaluations of neoadjuvant chemotherapy response are very important in clinical work and basic research. In this review, we will elaborate on evaluations of the efficacy of neoadjuvant chemotherapy in breast cancer and provide a clinical evaluation procedure for neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Huan Wang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
203
|
Mammatas LH, Zandvliet AS, Rovithi M, Honeywell RJ, Swart EL, Peters GJ, Menke-van der Houven van Oordt CW, Verheul HMW. Sorafenib administered using a high-dose, pulsatile regimen in patients with advanced solid malignancies: a phase I exposure escalation study. Cancer Chemother Pharmacol 2020; 85:931-940. [PMID: 32274565 PMCID: PMC7188706 DOI: 10.1007/s00280-020-04065-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/24/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND (Pre)clinical evidence is accumulating that intermittent exposure to increased doses of protein kinase inhibitors may improve their treatment benefit. In this phase I trial, the safety of high-dose, pulsatile sorafenib was studied. PATIENTS AND METHODS High-dose sorafenib was administered once weekly in exposure escalation cohorts according to a 3 + 3 design. Drug monitoring was performed in weeks 1-3 and doses were adjusted to achieve a predefined target plasma area under the curve (AUC)(0-12 h). The effect of low gastric pH on improving sorafenib exposure was investigated by intake of the acidic beverage cola. RESULTS Seventeen patients with advanced malignancies without standard treatment options were included. Once weekly, high-dose sorafenib exposure was escalated up to a target AUC(0-12 h) of 125-150 mg/L/h, achieving a twofold higher Cmax compared to standard continuous dosing. Dose-limiting toxicity was observed in three patients: grade 3 duodenal perforation (2800 mg sorafenib), grade 5 multiorgan failure (2800 mg sorafenib) and grade 5 biliary tract perforation (3600 mg sorafenib). The mean difference between observed and target AUC(0-12 h) was 45% (SD ± 56%) in week 1 using a fixed starting dose of sorafenib compared to 2% (SD ± 32%) in week 3 as a result of drug monitoring (P = 0.06). Dissolving sorafenib in cola, instead of water, did not improve sorafenib exposure. Clinical benefit with stable disease as the best response was observed in two patients. CONCLUSION Treatment with high-dose, once weekly sorafenib administration resulted in dose-limiting toxicity precluding dose escalation above the exposure cohort of 125-150 mg/L/h. Drug monitoring was a successful strategy to pursue a target exposure.
Collapse
Affiliation(s)
- L H Mammatas
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - A S Zandvliet
- Department of Clinical Pharmacology and Pharmacy, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - M Rovithi
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - R J Honeywell
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - E L Swart
- Department of Clinical Pharmacology and Pharmacy, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - G J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - C W Menke-van der Houven van Oordt
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - H M W Verheul
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VUmc University Medical Center Amsterdam, Amsterdam, The Netherlands.
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 8, Internal postal code 452, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
204
|
Chen HF, Wang WX, Xu CW, Huang LC, Li XF, Lan G, Zhai ZQ, Zhu YC, Du KQ, Lei L, Fang MY. A novel SOS1-ALK fusion variant in a patient with metastatic lung adenocarcinoma and a remarkable response to crizotinib. Lung Cancer 2020; 142:59-62. [PMID: 32114282 DOI: 10.1016/j.lungcan.2020.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Transforming anaplastic lymphoma kinase (ALK) gene rearrangements are well known as a unique subset of non-small cell lung cancer (NSCLC) with mutations other than EGFR. Currently, crizotinib is the standard first-line treatment for ALK-positive NSCLC. MATERIALS AND METHODS With advances in detection methods, more and more uncommon ALK fusion partners have been identified. Herein we present a novel SOS1-ALK fusion and the efficacy of crizotinib in an advanced NSCLC patient harboring this type of fusion. RESULTS A 52-year-old Chinese man had left upper lobe primary NSCLC and synchronous multiple lung metastases (cT2N3M1, stage IV). The ultrasound-guided fine-needle aspiration cytology of palpable left supraclavicular lymph nodes and the results of immunohistochemistry staining supported the diagnosis of metastatic lung adenocarcinoma. Using a next-generation sequencing assay (NGS), we showed that the tumor had a SOS1-ALK fusion which the breakpoints was (S2, A20) rather than other actionable mutations. Therefore, the patient received first-line crizotinib and experienced a remarkable tumor response and has tolerated crizotinib well until this writing. CONCLUSION Considering this rare SOS1-ALK fusion and remarkable response to an ALK-inhibitor, it is important to be aware of the presence of SOS1-ALK fusions in patients with advanced NSCLC to better guide targeted therapy. Precision methods, such as NGS for oncogenic alteration detection, should also be encouraged in clinical practice.
Collapse
Affiliation(s)
- Hua-Fei Chen
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - Wen-Xian Wang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou Zhejiang 310022, People's Republic of China
| | - Chun-Wei Xu
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou Fujian 350014, People's Republic of China.
| | - Li-Chao Huang
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - Xiao-Feng Li
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - Gang Lan
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - Zhan-Qiang Zhai
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - You-Cai Zhu
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - Kai-Qi Du
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital,The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang 314000, People's Republic of China
| | - Lei Lei
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou Zhejiang 310022, People's Republic of China.
| | - Mei-Yu Fang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou Zhejiang 310022, People's Republic of China
| |
Collapse
|
205
|
Wu D, Li X, Zhang X, Han F, Lu X, Liu L, Zhang J, Dong M, Yang H, Li H. Pharmacometabolomics Identifies 3-Hydroxyadipic Acid, d-Galactose, Lysophosphatidylcholine (P-16:0), and Tetradecenoyl-l-Carnitine as Potential Predictive Indicators of Gemcitabine Efficacy in Pancreatic Cancer Patients. Front Oncol 2020; 9:1524. [PMID: 32064236 PMCID: PMC7000527 DOI: 10.3389/fonc.2019.01524] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/18/2019] [Indexed: 12/28/2022] Open
Abstract
Gemcitabine (GEM)-based chemotherapy is the standard regimen for the treatment of pancreatic cancer (PC). However, chemoresistance is a major challenge in PC treatment. Reliable biomarkers are urgently needed to predict the response to GEM-based therapies. GEM-sensitive (GEM-S) and GEM-resistant (GEM-R) pancreatic carcinoma xenograft models were established, and GEM monotherapy and GEM plus nanoparticle albumin-bound paclitaxel (nab-PTX) doublet therapy were administered to GEM-S/R tumor-bearing mice. Metabolomic mass spectrometry (MS) analysis of serum, liver, and tumor samples was performed using an ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometer. The results showed that both GEM monotherapy and combination therapy significantly inhibited the tumor growth in GEM-S subgroup. However, in the GEM-R subgroup, tumor growth was not significantly inhibited by GEM monotherapy, but was significantly suppressed by GEM combination therapy. Metabolic profiling analysis by hierarchical cluster analysis and partial least squares discriminant analysis showed that the differences in metabolites were most significant in serum of three types of samples in the GEM-S/R subgroups, regardless of the administration of GEM monotherapy or combination therapy. The differential metabolite analysis of serum samples revealed 38 and 26 differential metabolites between the GEM-R and GEM-S subgroups treated with GEM monotherapy or combination therapy, and four common discriminating metabolites were investigated: 3-hydroxyadipic acid, d-galactose, lysophosphatidylcholine (LysoPC) (P-16:0), and tetradecenoyl-l-carnitine. The relative amounts of the four metabolites changed significantly and consistently after GEM monotherapy or combination therapy. The levels of these four metabolites were significantly different in the GEM-S and GEM-R pancreatic carcinoma xenograft models; thus, these metabolites could be effective predictive indicators of the efficacy of chemotherapy in PC patients, regardless of the administration of GEM alone or GEM plus nab-PTX.
Collapse
Affiliation(s)
- Dongyuan Wu
- Department of Biochemistry and Molecular Biology, Basic Medical Science College, Harbin Medical University, Harbin, China.,Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xinyuan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaohan Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Fang Han
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xin Lu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Lei Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Junsheng Zhang
- College of Basic Medicine, Harbin Medical University, Harbin, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Hui Li
- Department of Biochemistry and Molecular Biology, Basic Medical Science College, Harbin Medical University, Harbin, China
| |
Collapse
|
206
|
Inhibiting WNT and NOTCH in renal cancer stem cells and the implications for human patients. Nat Commun 2020; 11:929. [PMID: 32066735 PMCID: PMC7026425 DOI: 10.1038/s41467-020-14700-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/21/2020] [Indexed: 02/08/2023] Open
Abstract
Current treatments for clear cell renal cell cancer (ccRCC) are insufficient because two-thirds of patients with metastases progress within two years. Here we report the identification and characterization of a cancer stem cell (CSC) population in ccRCC. CSCs are quantitatively correlated with tumor aggressiveness and metastasis. Transcriptional profiling and single cell sequencing reveal that these CSCs exhibit an activation of WNT and NOTCH signaling. A significant obstacle to the development of rational treatments has been the discrepancy between model systems and the in vivo situation of patients. To address this, we use CSCs to establish non-adherent sphere cultures, 3D tumor organoids, and xenografts. Treatment with WNT and NOTCH inhibitors blocks the proliferation and self-renewal of CSCs in sphere cultures and organoids, and impairs tumor growth in patient-derived xenografts in mice. These findings suggest that our approach is a promising route towards the development of personalized treatments for individual patients. Cancer stem cells are thought to be largely resistant to treatment and can be responsible for tumour recurrence. Here, using renal cancer organoids, self-renewing sphere cultures and PDX from patients, the authors show that the proliferation of stem cells within organoids, PDX and spheres can be blocked by the concomitant inhibition of the NOTCH and WNT pathways.
Collapse
|
207
|
Rau KM, Liu CT, Hsiao YC, Hsiao KY, Wang TM, Hung WS, Su YL, Liu WC, Wang CH, Hsu HL, Chuang PH, Cheng JC, Tseng CP. Sequential Circulating Tumor Cell Counts in Patients with Locally Advanced or Metastatic Hepatocellular Carcinoma: Monitoring the Treatment Response. J Clin Med 2020; 9:E188. [PMID: 32071283 PMCID: PMC7019972 DOI: 10.3390/jcm9010188] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/05/2020] [Accepted: 01/08/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is among the most common causes of cancer death in men. Whether or not a longitudinal follow-up of circulating tumor cells (CTCs) before and at different time points during systemic/targeted therapy is useful for monitoring the treatment response of patients with locally advanced or metastatic HCC has been evaluated in this study. Blood samples (n = 104) were obtained from patients with locally advanced or metastatic HCC (n = 30) for the enrichment of CTCs by a negative selection method. Analysis of the blood samples from patients with defined disease status (n = 81) revealed that those with progressive disease (PD, n = 37) had significantly higher CTC counts compared to those with a partial response (PR) or stable disease (SD; n = 44 for PR + SD, p = 0.0002). The median CTC count for patients with PD and for patients with PR and SD was 50 (interquartile range 21-139) and 15 (interquartile range 4-41) cells/mL of blood, respectively. A longitudinal analysis of patients (n = 17) after a series of blood collections demonstrated that a change in the CTC count correlated with the patient treatment response in most of the cases and was particularly useful for monitoring patients without elevated serum alpha-fetoprotein (AFP) levels. Sequential CTC enumeration during treatment can supplement standard medical tests and benefit the management of patients with locally advanced or metastatic HCC, in particular for the AFP-low cases.
Collapse
Affiliation(s)
- Kun-Ming Rau
- Department of Hematology-Oncology, E-Da Cancer Hospital, Kaohsiung 824, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
| | - Chien-Ting Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-T.L.); (K.-Y.H.); (Y.-L.S.); (W.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Yu-Chiao Hsiao
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.H.); (T.-M.W.); (W.-S.H.); (H.-L.H.)
| | - Kai-Yin Hsiao
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-T.L.); (K.-Y.H.); (Y.-L.S.); (W.-C.L.)
| | - Tzu-Min Wang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.H.); (T.-M.W.); (W.-S.H.); (H.-L.H.)
| | - Wei-Shan Hung
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.H.); (T.-M.W.); (W.-S.H.); (H.-L.H.)
| | - Yu-Li Su
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-T.L.); (K.-Y.H.); (Y.-L.S.); (W.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Wei-Ching Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-T.L.); (K.-Y.H.); (Y.-L.S.); (W.-C.L.)
| | - Cheng-Hsu Wang
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Division of Hematology/Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Hsueh-Ling Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.H.); (T.-M.W.); (W.-S.H.); (H.-L.H.)
| | - Po-Heng Chuang
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan;
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan;
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.H.); (T.-M.W.); (W.-S.H.); (H.-L.H.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 333, Taiwan
- Department of Laboratory Medicine, Linko Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
208
|
Gorospe L, Pacios-Blanco RE, Garrido-López P. The Importance of Imaging Studies in the Assessment of Response to Immunotherapy in Lung Cancer. Arch Bronconeumol 2020; 56:380-389. [PMID: 31898993 DOI: 10.1016/j.arbres.2019.10.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
Immunotherapy (particularly immune checkpoint inhibitors) in the treatment of patients with lung cancer has aroused great interest in recent years, revolutionized the management of patients with locally advanced/metastatic disease, and given hope to both patients and treating physicians. These drugs, in combination or in monotherapy, have become the standard treatment for many patients with lung cancer, and their use is expected to increase significantly in the near future. In this article, we will review the growing importance of imaging techniques in the evaluation of therapeutic response to immunotherapy in lung cancer patients, with emphasis on the new specific radiological criteria on response to immunotherapy, atypical radiological responses (pseudoprogresion, dissociative responses, hyperprogresion), and the main radiological manifestations of adverse events associated with immunotherapy (sarcoid reactions, pulmonary toxicities, etc.). Pulmonologists must be familiar not only with atypical radiological responses to immunotherapy and their prognostic implications, but also with their effects and the new radiological criteria of response to assess treatment response. In this study, we will address key concepts such as "pseudoprogresion", "paradoxical response", "hyperprogresion", or "unconfirmed progression", and their significance in the management of patients with lung cancer treated with immunotherapy.
Collapse
Affiliation(s)
- Luis Gorospe
- Servicio de Radiodiagnóstico, Hospital Universitario Ramón y Cajal, Madrid, España.
| | | | - Pilar Garrido-López
- Servicio de Oncología Médica, Hospital Universitario Ramón y Cajal, Madrid, España
| |
Collapse
|
209
|
Ameli S, Shaghaghi M, Kamel IR, Zaheer A. Therapy Response Imaging in Hepatobiliary and Pancreatic Malignancies. MEDICAL RADIOLOGY 2020:117-137. [DOI: 10.1007/978-3-030-31171-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
210
|
Zhou X, Liu M, Ren Q, Zhu W, Wang Y, Chen H, Chen J. Oral and injectable Marsdenia tenacissima extract (MTE) as adjuvant therapy to chemotherapy for gastric cancer: a systematic review. Altern Ther Health Med 2019; 19:366. [PMID: 31830977 PMCID: PMC6909592 DOI: 10.1186/s12906-019-2779-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022]
Abstract
Background Marsdenia tenacissima extract (MTE) is a phytochemical widely used as complementary therapy in cancer care. This systematic review was conducted to investigate the anticancer and detoxification effects of MTE, as an adjuvant therapy to chemotherapy, for treating gastric cancer. Methods Ten databases were searched to identify randomized controlled trials (RCTs) comparing oral or injectable MTE plus chemotherapy versus chemotherapy alone for treating gastric cancer up to May 1, 2019. In meta-analyses, proportional odds ratios (PORs) with 95% confidence intervals (CIs) were pooled for the ordinal outcomes using the generalized linear model, and risk ratios (RRs) with 95% CIs were pooled for dichotomous outcomes using the Mantel-Haenszel method. Results Seventeen RCTs with 1329 individuals were included, with a moderate to high risk of selection and performance bias. Compared to chemotherapy alone, MTE adjuvant therapy significantly improved the response to anticancer treatment (POR 2.01, 95% CI 1.60–2.53) and patients’ performance status (POR 3.15, 95% CI 2.22–4.48) and reduce the incidences of chemotherapy-induced leukopenia (RR 0.66, 95% CI 0.56–0.78), thrombocytopenia (RR 0.64, 95% CI 0.48–0.86), anemia (RR 0.89, 95% CI 0.72–1.10), nausea/vomiting (RR 0.79, 95% CI 0.69–0.91), hepatic injury (RR 0.77, 95% CI 0.61–0.96), and peripheral neurotoxicity (RR 0.77, 95% CI 0.59–1.01). However, MTE did not significantly alleviate anemia, diarrhea, constipation, kidney injury, and oral mucosal lesions after chemotherapy. Incidence of nausea/vomiting was lower in patients receiving oral MTE than those receiving injectable MTE (RR 0.47 vs. 0.82, interaction P = 0.04). Heterogeneity was generally low among these outcomes. Three out of five RCTs that reported survival data supported the effects of MTE for prolonging progression-free and/or overall survival. No studies reported safety outcomes of MTE. Conclusions The current evidence with limitations of risk of selection and performance bias suggests that MTE, as an adjuvant therapy to chemotherapy, is effective for inhibiting cancer growth and reducing incidences of multiple chemotherapy side effects. Oral MTE may be a better choice. Uncertainty remains regarding the effects of MTE on survival endpoints and the subgroup differences between acute and chronic use of MTE and between different chemotherapy regimens.
Collapse
|
211
|
Lin J, Yang X, Xie Y, Zhao H. Unleashing The Potential Of Multiple-Targeted Therapies In Hepatobiliary Cancers: Two Cases, Cocktail Therapy With Nine Molecular Targeted Agents And Long-Lasting Survival. Onco Targets Ther 2019; 12:9941-9945. [PMID: 31819490 PMCID: PMC6875264 DOI: 10.2147/ott.s223568] [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: 07/17/2019] [Accepted: 11/02/2019] [Indexed: 11/23/2022] Open
Abstract
The mortality of hepatobiliary cancer still stays high around the world, with disappointing treatment status at advanced stage. Genomic analysis has identified the global spectrum of alterations in liver cancer, but the most common mutations are not actionable, meanwhile lacking potent molecular-targeted drugs for specific oncogenic drivers. Multiple-targeted drugs which mainly inhibited tumors’ angiogenesis, such as sorafenib and lenvatinib, improved survival time for patients with advanced hepatocellular carcinoma, although the median overall survival remains 1–1.5 years. It is still ambiguous for the precise biomarkers of multiple-targeted drugs utilized in hepatobiliary cancer. Herein, we reported two observational patients who were sequentially treated by various targeted drugs and obtained lasting overall survival time. Both the two patients weekly switched targeted drugs according to the changes of tumor markers. These two cases would get us thinking: what is the underlying mechanism of molecular-targeted drugs in hepatobiliary tumors; and whether a “drugs screening model” during real-time treatment could be achieved through the assistance from sensitive and specific tumor markers.
Collapse
Affiliation(s)
- Jianzhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, People's Republic of China
| | - Xiaobo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, People's Republic of China
| | - Yuan Xie
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, People's Republic of China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, People's Republic of China
| |
Collapse
|
212
|
Hufnagl C, Leisch M, Weiss L, Melchardt T, Moik M, Asslaber D, Roland G, Steininger P, Meissnitzer T, Neureiter D, Greil R, Egle A. Evaluation of circulating cell-free DNA as a molecular monitoring tool in patients with metastatic cancer. Oncol Lett 2019; 19:1551-1558. [PMID: 31966080 DOI: 10.3892/ol.2019.11192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 08/06/2019] [Indexed: 01/16/2023] Open
Abstract
The clinical decisions made when treating patients with metastatic cancer require knowledge of the current tumor extent and response to therapy. For the majority of solid tumors, a response assessment, which is based on imaging, is used to guide these decisions. However, measuring serum protein biomarkers (i.e. tumor markers) may be of additional use. Furthermore, tumor markers exhibit variable specificity and sensitivity and cannot therefore be solely relied upon when making decisions regarding cancer treatment. Therefore, there is a clinical requirement for the identification of specific, sensitive and quantitative biomarkers. In recent years, circulating cell-free DNA (cfDNA) and mutation-specific circulating cell-free tumor DNA (cftDNA) have been identified as novel potential biomarkers. In the current study, cfDNA and cftDNA were compared using imaging-based staging and current tumor markers in 15 patients with metastatic colorectal, pancreatic or breast cancer. These patients were treated at the Third Medical Department of Paracelsus Medical University Salzburg (Austria). The results of the current study demonstrated a statistically significant correlation between the concentration changes of cfDNA and cftDNA and response to treatment, which was assessed by imaging. A correlation was not indicated with current clinically used tumor markers, including carcinoembryonic antigen, carcinoma antigen 15-3 and carcinoma antigen 19-9. The present study also indicated a correlation between cfDNA and cftDNA and the tumor volume of metastatic lesions, which was not observed with the current clinically used tumor markers. In conclusion, cfDNA and cftDNA exhibit the potential to become novel biomarkers for the response assessment following cancer treatment, and may serve as a tool for the estimation of tumor volume. The current study further supports the increasingly important role of cfDNA and cftDNA as new monitoring tools for use during cancer therapy.
Collapse
Affiliation(s)
- Clemens Hufnagl
- Institute of Pathology, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria
| | - Michael Leisch
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Lukas Weiss
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Thomas Melchardt
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Martin Moik
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Daniela Asslaber
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Geisberger Roland
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Philipp Steininger
- Institute for Research and Development on Advanced Radiation Technologies, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria
| | - Thomas Meissnitzer
- Institute of Radiology, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria
| | - Richard Greil
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| | - Alexander Egle
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, A-5020 Salzburg, Austria.,Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, A-5020 Salzburg, Austria.,Cancer Cluster Salzburg, A-5020 Salzburg, Austria
| |
Collapse
|
213
|
Bogani G, Vinti D, Murgia F, Chiappa V, Leone Roberti Maggiore U, Martinelli F, Ditto A, Raspagliesi F. Burden of lymphatic disease predicts efficacy of adjuvant radiation and chemotherapy in FIGO 2018 stage IIICp cervical cancer. Int J Gynecol Cancer 2019; 29:1355-1360. [PMID: 31422352 DOI: 10.1136/ijgc-2019-000669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE Nodal involvement is one of the most important prognostic factors in cervical cancer patients. We aimed to assess the prognostic role in relation to the burden of nodal disease in stage IIICp cervical cancer. METHODS Data on all consecutive patients diagnosed with cervical cancer undergoing primary surgery (radical hysterectomy plus lymphadenectomy) or neoadjuvant chemotherapy followed by radical hysterectomy plus lymphadenectomy, between January 1980 and December 2017, were collected in a dedicated database. Exclusion criteria were: (1) consent withdrawal; (2) synchronous malignancies (within 5 years). Survival outcomes were assessed using Kaplan-Meier and Cox models. RESULTS Overall, 177 (14.1%) of 1257 patients with cervical cancer were diagnosed with positive lymph nodes. After a median follow-up of 58 (range 4-175) months, 66 (37.3%) and 37 (20.9%) patients developed recurrent disease and died of disease, respectively. Via multivariate analysis, positive para-aortic nodes (HR 2.62, 95% CI 1.12 to 6.11; p=0.025) and the number of positive nodes (HR 1.06, 95% CI 1.02 to 1.11; p=0.002) correlated with worse disease-free survival. Furthermore, the number of positive nodes (HR 1.06, 95% CI 1.01 to 1.12; p=0.021) correlated with worse overall survival. Number of positive nodes (1, 2 or ≥3) strongly correlated with both disease-free survival (p<0.001, log-rank test) and overall survival (p=0.001, log-rank test). Focusing on patients receiving adjuvant radiation and chemotherapy, the number of positive lymph nodes was associated with response to treatment (p<0.001). Median disease-free survival was 100, 42, and 12 months for patients with one, two, or three or more positive lymph node(s), respectively (p<0.001, log-rank test). CONCLUSIONS In stage IIICp cervical cancer, adjuvant radiation and chemotherapy provides adequate overall survival in patients diagnosed with only one metastatic node, while survival outcomes are poor in patients with two or more metastatic nodes. This highlights the need for innovative treatments in patients with a high burden of lymphatic disease.
Collapse
Affiliation(s)
- Giorgio Bogani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Daniele Vinti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | | | | | | | - Antonino Ditto
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | |
Collapse
|
214
|
Lalchandani UR, Sahai V, Hersberger K, Francis IR, Wasnik AP. A Radiologist's Guide to Response Evaluation Criteria in Solid Tumors. Curr Probl Diagn Radiol 2019; 48:576-585. [DOI: 10.1067/j.cpradiol.2018.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 01/09/2023]
|
215
|
Omballi M, Fernandez-Bussy S, Patel PP, Jantz MA, Becnel D, Patel NM, Mehta HJ. Surveillance Imaging After Curative Intent Therapy for Lung Cancer. Semin Roentgenol 2019; 55:60-69. [PMID: 31964482 DOI: 10.1053/j.ro.2019.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Mohamed Omballi
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | | | - Priya P Patel
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Michael A Jantz
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - David Becnel
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - Neal M Patel
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| | - Hiren J Mehta
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL.
| |
Collapse
|
216
|
Liu G, Wang C, He Y, E M. Application effect of apatinib in patients with failure of standard treatment for advanced malignant tumours. BMC Pharmacol Toxicol 2019; 20:61. [PMID: 31661009 PMCID: PMC6819520 DOI: 10.1186/s40360-019-0362-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/02/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In recent years, targeted therapy has received widespread attention. Among these therapies, anti-angiogenic targeted drugs have become one of the hotspots of research. Apatinib is a novel oral small molecule anti-angiogenic agent that has been clinically tested in a variety of solid tumours. The aim of this study was to investigate the efficacy of apatinib in patients with advanced malignant tumours and failure of standard therapy. METHODS We collected 41 patients with advanced malignant tumours in our department; all tumours were pathologically confirmed as malignant. All patients received apatinib after failure of standard therapy: 500 mg/dose, one dose/d, orally 30 min after a meal, until progressive disease or intolerable adverse reactions occurred. When there was a second- or third-degree adverse reaction associated with apatinib during treatment, apatinib treatment could be suspended or reduced to 250 mg/dose. Clinical efficacy and progression-free survival were assessed according to RECIST1.1, and adverse reactions were observed. RESULTS Efficacy assessment was available for 31 patients with a median progression-free survival time of 2.66 months; the objective response rate and disease control rates were 16.1 and 64.5%, respectively. The disease control rates of the patients with lower Eastern Cooperative Oncology Group scores (1-2 points) and with fewer metastatic sites (< 3 sites) were higher than those of the patients with higher scores (3 points) and with more metastatic sites (≥3 sites), respectively (all P < 0.05). The most common adverse reactions were hypertension, neutropenia and hand-foot syndrome. CONCLUSION For patients with advanced malignant tumours with failure of standard therapy, administration of apatinib can still result in good efficacy. The efficacy of apatinib is better in patients with a higher performance status and lower degree of tumour progression.
Collapse
Affiliation(s)
- Guohui Liu
- Department of Radiation Oncology, The Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Chunbo Wang
- Department of Radiation Oncology, The Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yunlong He
- Department of Radiation Oncology, The Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Mingyan E
- Department of Radiation Oncology, The Harbin Medical University Cancer Hospital, Harbin, 150040, China.
| |
Collapse
|
217
|
Fugazzola L, Elisei R, Fuhrer D, Jarzab B, Leboulleux S, Newbold K, Smit J. 2019 European Thyroid Association Guidelines for the Treatment and Follow-Up of Advanced Radioiodine-Refractory Thyroid Cancer. Eur Thyroid J 2019; 8:227-245. [PMID: 31768334 PMCID: PMC6873012 DOI: 10.1159/000502229] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/19/2019] [Indexed: 01/03/2023] Open
Abstract
The vast majority of thyroid cancers of follicular origin (TC) have a very favourable outcome, but 5-10% of cases will develop metastatic disease. Around 60-70% of this subset, hence less than 5% of all patients with TC, will become radioiodine refractory (RAI-R), with a significant negative impact on prognosis and a mean life expectancy of 3-5 years. Since no European expert consensus or guidance for this challenging condition is currently available, a task force of TC experts was nominated by the European Thyroid Association (ETA) to prepare this document based on the principles of clinical evidence. The task force started to work in September 2018 and after several revision rounds, prepared a list of recommendations to support the treatment and follow-up of patients with advanced TC. Criteria for advanced RAI-R TC were proposed, and the most appropriate diagnostic tools and the local, systemic and palliative treatments are described. Systemic therapy with multikinase inhibitors is fully discussed, including recommendations on how to start it and at which dosage, on the duration of treatment, and on the management of side effects. The appropriate relationship between the specialist and the patient/family as well as ethical issues are covered. Based on the available studies and on personal experience, the experts provided 39 recommendations aimed to improve the management of advanced RAI-R TCs. Above all of them is the indication to treat and follow these patients in a specialized setting which allows the interaction between several specialists in a multidisciplinary team.
Collapse
Affiliation(s)
- Laura Fugazzola
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Rossella Elisei
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Dagmar Fuhrer
- Department of Endocrinology, Diabetes and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Barbara Jarzab
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Sophie Leboulleux
- Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy and Université Paris Saclay, Villejuif, France
| | - Kate Newbold
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| | - Jan Smit
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
218
|
Alipour R, Azad A, Hofman MS. Guiding management of therapy in prostate cancer: time to switch from conventional imaging to PSMA PET? Ther Adv Med Oncol 2019; 11:1758835919876828. [PMID: 31565073 PMCID: PMC6755643 DOI: 10.1177/1758835919876828] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/27/2019] [Indexed: 11/17/2022] Open
Abstract
Radiolabelled small molecules for imaging prostate cancer have rapidly emerged
over the last few years with gallium-68-labelled
prostate-specific-membrane-antigen-11 (68Ga-PSMA11), the most widely
used. However, the current evidence-based guidelines for management of prostate
cancer were established using computed tomography (CT), magnetic resonance
imaging (MRI) and bone scan, despite their limitations.
Prostate-specific-membrane antigen (PSMA) positron-emission tomography (PET)/CT,
however, has higher sensitivity and specificity and can lead to both upstaging
and downstaging and subsequent changes in management of prostate cancer. The
literature for PSMA PET/CT is mostly in the setting of biochemical recurrence
and primary staging of intermediate-to-high-risk prostate cancer. Preliminary
studies also suggest that there may be a role in nonmetastatic
castrate-resistant prostate cancer (nmCRPC) and possibly response to therapy.
Despite high sensitivity and specificity, PSMA PET/CT as a single modality for
staging advanced prostate cancer is suboptimal, given the low PSMA expression in
this subgroup and the complementary role of fluorodeoxyglucose (FDG) PET/CT is
required. This is also true in early-stage prostate adenocarcinoma with
neuroendocrine differentiation or small-/large-cell neuroendocrine tumours of
the prostate. Lack of a globally accepted standardized reporting system for PSMA
PET/CT is a current limitation. This is essential to pave the way to
incorporating this invaluable molecular imaging modality in clinical trials to
assess its impact on outcome, particularly when upstaging or downstaging
conventionally imaged disease. This would then lead to recognition by healthcare
providers, incorporation into guidelines for management of prostate cancer and
routine use in clinical practice.
Collapse
Affiliation(s)
- Ramin Alipour
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Level 5, 305 Grattan Street, Melbourne, VIC 3000, Australia
| | - Arun Azad
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael S Hofman
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
219
|
Challapalli A, Pearson S, Mitra AV, Coe M, Thomson A, Elliott T, Kirkbride P, Pickering L, Kirk H, Foulstone E, Evans H, Bravo A, Bahl AK. A phase II trial of cabazitaxel as second line chemotherapy in relapsed locally advanced and/or metastatic carcinoma of the penis. J Int Med Res 2019; 47:4664-4672. [PMID: 31502503 PMCID: PMC6833416 DOI: 10.1177/0300060519863546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objectives To assess the efficacy and tolerability of cabazitaxel in relapsed penile cancer. Methods This Phase II single-arm trial was designed to recruit 17 patients with relapsed penile cancer. The primary endpoint was objective (complete + partial) response rate (ORR; Response Evaluation Criteria in Solid Tumours [RECIST] v1.1). Treatment comprised six 21-day cycles of cabazitaxel with restaging after cycles 2 and 4. The planned interim analysis was based upon the premise that if none of the first nine patients achieved ORR, trial would be stopped (α = 0.05, Simon’s 2-stage design). Results Nine patients were recruited from four UK centres between December 2014 and August 2016. The median age was 61 (range, 27–73.6) years, and seven patients had metastases. Patients received a median of two chemotherapy cycles (range, 2–5). None of the nine patients achieved ORR and the trial was stopped. Cabazitaxel was well tolerated with no dose reductions or delays. Three patients had grade 3/4 adverse events (anaemia, vomiting, or neutropenic sepsis). The median progression-free and overall survival were 1.3 and 5.6 months, respectively. Conclusions The trial did not reach the threshold for further continuation of single-agent cabazitaxel. However, the observed tolerability profile supports its further investigation in combination with other agents to improve patient outcomes.
Collapse
Affiliation(s)
| | - Sylvia Pearson
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| | - Anita V Mitra
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Marc Coe
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| | | | - Tony Elliott
- The Christie NHS Foundation Trust, Manchester, UK
| | - Peter Kirkbride
- The Clatterbridge Cancer Centre NHS Foundation Trust, The Wirral, UK
| | - Lisa Pickering
- St Georges University Hospitals NHS Foundation Trust, London, UK
| | - Hannah Kirk
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| | - Emily Foulstone
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| | - Heidi Evans
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| | - Alicia Bravo
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| | - Amit K Bahl
- Department of Clinical Oncology, Bristol Cancer Institute, Bristol, UK
| |
Collapse
|
220
|
Duran-Moreno J, Kampoli K, Kapetanakis EI, Mademli M, Koufopoulos N, Foukas PG, Kostopanagiotou K, Tomos P, Koumarianou A. Pericardial Synovial Sarcoma: Case Report, Literature Review and Pooled Analysis. In Vivo 2019; 33:1531-1538. [PMID: 31471401 PMCID: PMC6754991 DOI: 10.21873/invivo.11633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pericardial synovial sarcomas (PSS) are very rare tumors, with dismal prognosis and limited data. We describe the clinical features and identify prognostic factors of primary PSS. CASE REPORT We describe the case of a 56-year-old male patient with PSS managed by the multidisciplinary team of thoracic oncology. The therapeutic plan comprised surgery, chemotherapy, stereotactic radiosurgery and targeted therapy, with excellent results. MATERIALS AND METHODS Data from 37 cases reported in English during the past 20 years were gathered and analyzed. PSS was found to occur at a mean age of 36±17.082 (range=13-67) years. Survival analysis was performed on 20 cases with follow-up of at least 6 months. CONCLUSION Only complete resection of the tumor seems to be an independent prognostic factor. To our knowledge, this is the first report on the safety and effectivity of pazopanib in PSS and may provide guidance for similar cases in the future.
Collapse
Affiliation(s)
- Jose Duran-Moreno
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Kampoli
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil I Kapetanakis
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Mademli
- Second Department of Radiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Koufopoulos
- Second Department of Pathology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis G Foukas
- Second Department of Pathology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Kostopanagiotou
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis Tomos
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
221
|
deSouza NM, Achten E, Alberich-Bayarri A, Bamberg F, Boellaard R, Clément O, Fournier L, Gallagher F, Golay X, Heussel CP, Jackson EF, Manniesing R, Mayerhofer ME, Neri E, O'Connor J, Oguz KK, Persson A, Smits M, van Beek EJR, Zech CJ. Validated imaging biomarkers as decision-making tools in clinical trials and routine practice: current status and recommendations from the EIBALL* subcommittee of the European Society of Radiology (ESR). Insights Imaging 2019; 10:87. [PMID: 31468205 PMCID: PMC6715762 DOI: 10.1186/s13244-019-0764-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
Observer-driven pattern recognition is the standard for interpretation of medical images. To achieve global parity in interpretation, semi-quantitative scoring systems have been developed based on observer assessments; these are widely used in scoring coronary artery disease, the arthritides and neurological conditions and for indicating the likelihood of malignancy. However, in an era of machine learning and artificial intelligence, it is increasingly desirable that we extract quantitative biomarkers from medical images that inform on disease detection, characterisation, monitoring and assessment of response to treatment. Quantitation has the potential to provide objective decision-support tools in the management pathway of patients. Despite this, the quantitative potential of imaging remains under-exploited because of variability of the measurement, lack of harmonised systems for data acquisition and analysis, and crucially, a paucity of evidence on how such quantitation potentially affects clinical decision-making and patient outcome. This article reviews the current evidence for the use of semi-quantitative and quantitative biomarkers in clinical settings at various stages of the disease pathway including diagnosis, staging and prognosis, as well as predicting and detecting treatment response. It critically appraises current practice and sets out recommendations for using imaging objectively to drive patient management decisions.
Collapse
Affiliation(s)
- Nandita M deSouza
- Cancer Research UK Imaging Centre, The Institute of Cancer Research and The Royal Marsden Hospital, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | | | | | - Fabian Bamberg
- Department of Radiology, University of Freiburg, Freiburg im Breisgau, Germany
| | | | | | | | | | | | - Claus Peter Heussel
- Universitätsklinik Heidelberg, Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
| | - Edward F Jackson
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rashindra Manniesing
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, 6525, GA, Nijmegen, The Netherlands
| | | | - Emanuele Neri
- Department of Translational Research, University of Pisa, Pisa, Italy
| | - James O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | | | | | - Marion Smits
- Department of Radiology and Nuclear Medicine (Ne-515), Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - Edwin J R van Beek
- Edinburgh Imaging, Queen's Medical Research Institute, Edinburgh Bioquarter, 47 Little France Crescent, Edinburgh, UK
| | - Christoph J Zech
- University Hospital Basel, Radiology and Nuclear Medicine, University of Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| |
Collapse
|
222
|
AI-based applications in hybrid imaging: how to build smart and truly multi-parametric decision models for radiomics. Eur J Nucl Med Mol Imaging 2019; 46:2673-2699. [PMID: 31292700 DOI: 10.1007/s00259-019-04414-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The quantitative imaging features (radiomics) that can be obtained from the different modalities of current-generation hybrid imaging can give complementary information with regard to the tumour environment, as they measure different morphologic and functional imaging properties. These multi-parametric image descriptors can be combined with artificial intelligence applications into predictive models. It is now the time for hybrid PET/CT and PET/MRI to take the advantage offered by radiomics to assess the added clinical benefit of using multi-parametric models for the personalized diagnosis and prognosis of different disease phenotypes. OBJECTIVE The aim of the paper is to provide an overview of current challenges and available solutions to translate radiomics into hybrid PET-CT and PET-MRI imaging for a smart and truly multi-parametric decision model.
Collapse
|
223
|
Limited-Stage Small Cell Lung Cancer: Is Prophylactic Cranial Irradiation Necessary? Pract Radiat Oncol 2019; 9:e599-e607. [PMID: 31271904 DOI: 10.1016/j.prro.2019.06.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Prophylactic cranial irradiation (PCI) reduces the incidence of brain metastases in patients with limited stage small cell lung cancer (LS-SCLC). However, PCI is associated with neurotoxicity. Previous studies have not consistently used pretreatment magnetic resonance imaging. Modern imaging improvements continue to enhance early metastasis detection, potentially decreasing the utility of PCI. We sought to determine whether PCI was associated with improved outcomes in LS-SCLC patients with modern imaging. METHODS AND MATERIALS We identified LS-SCLC patients with no intracranial disease who were treated between 2007 and 2018. Kaplan-Meier estimates of overall survival (OS) and progression-free survival (PFS) were calculated and multivariate Cox proportional hazards models were generated. The cumulative incidence of brain metastases was estimated using competing risks methodology. RESULTS Ninety-two patients were identified without intracranial disease at initial staging, 39 of whom received PCI. Median follow-up was 56.7 months. The median OS for the cohort was 35.5 months (95% CI, 25.8-49.3), and median PFS was 19.1 months (95% CI, 12.3-30.5). Median OS with PCI versus observation was 37.9 months (95% CI, 31.8-not reached) versus 30.5 months (95% CI, 14.6-56.1; P = .07), whereas median PFS was 26.3 months (95% CI 19.1-not reached) versus 12.3 months (95% CI, 8.5-30.5; P = .02), respectively. Overall, at 2 years, the cumulative incidence of brain metastases was 10% with PCI and 29% without; this increased to 32% and 29% by 4 years (P = .66). In those patients who had negative magnetic resonance imaging of the brain after completing initial treatment, the 1-year cumulative incidence of brain metastasis was not significantly different at 8% versus 11% (P = .46) respectively. Both PCI and treatment response were independent predictors for PFS on multivariate analysis. Stratified by disease response, patients with a complete response did not benefit from PCI (P = .50), whereas those with partial response or stable disease experienced improved PFS (P = .01). CONCLUSIONS Overall, PCI was associated with improved PFS and reduced early incidence of brain metastases. Patients achieving a complete response to initial therapy did not experience a PFS benefit with PCI. This may indicate that subsets of LS-SCLC patients can potentially be spared from PCI in the era of modern imaging.
Collapse
|
224
|
Faraji F, Gaba RC. Radiologic Modalities and Response Assessment Schemes for Clinical and Preclinical Oncology Imaging. Front Oncol 2019; 9:471. [PMID: 31214510 PMCID: PMC6558006 DOI: 10.3389/fonc.2019.00471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 05/16/2019] [Indexed: 11/29/2022] Open
Abstract
Clinical drug trials for oncology have resulted in universal protocols for medical imaging in order to standardize protocols for image procurement, radiologic interpretation, and therapeutic response assessment. In recent years, there has been increasing interest in using large animal models to study oncologic disease, though few standards currently exist for imaging of large animal models. This article briefly reviews medical imaging modalities, the current state-of-the-art in radiologic diagnostic criteria and response assessment schemes for evaluating therapeutic response and disease progression, and translation of radiologic imaging protocols and standards to large animal models of malignant disease.
Collapse
Affiliation(s)
- Farshid Faraji
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Ron C Gaba
- Department of Radiology, University of Illinois Health, Chicago, IL, United States
| |
Collapse
|
225
|
Faehling M, Kopp M, Schwenk B, Fallscheer S, Kramberg S, Eckert R. Immuno-Oncological Treatment and Tumor Mass in Non-Small Cell Lung Cancer: Case-Control Analysis of Overall Survival in Routine Clinical Practice. Oncology 2019; 97:228-235. [DOI: 10.1159/000500885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/26/2019] [Indexed: 11/19/2022]
|
226
|
Hess LM, Brnabic A, Mason O, Lee P, Barker S. Relationship between Progression-free Survival and Overall Survival in Randomized Clinical Trials of Targeted and Biologic Agents in Oncology. J Cancer 2019; 10:3717-3727. [PMID: 31333789 PMCID: PMC6636299 DOI: 10.7150/jca.32205] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/15/2019] [Indexed: 12/20/2022] Open
Abstract
Introduction: With a gap in a full understanding of the mechanisms by which survival is extended for patients with cancer who are treated with novel biologic and targeted agents, there is the risk that discordant progression-free and overall survival outcomes are observed due to poor clinical trial design or biases in the interpretation of data. This study was designed to examine the role of study quality and design on the outcomes observed with biologic and targeted agents. Methods: A review of studies in clinicaltrials.gov supplemented with a literature review in OVID Medline was conducted to identify all randomized trials of a biologic/targeted agent versus a non-biologic/targeted comparator in oncology that report both median overall and progression-free survival outcomes. Details of the study, design, population, drugs, and outcomes were extracted. Study quality was evaluated using the PEDro scale. Data were summarized using SPSS 22.0.0.0. Results: A total of 192 unique studies of 206 pairwise comparisons between a biologic/targeted and comparator were identified. The average absolute magnitude of post-progression survival (difference between OS and PFS) was 9.7 months for biologic/targeted therapy and 9.8 for the comparator. A total of 64 comparisons (31.1%) showed an increase in OS and decrease in PFS, or vice versa, and 25 (12.1%) showed a magnitude of more than 4 months difference between the delta of OS and delta of PFS between the biologic/targeted and comparator arms. Average study quality was high overall (7.7/10), and was comparable for studies with directional differences (7.2/10) as well as for those with the greatest magnitude in post-progression survival (7.4/10). Conclusion: This review and analysis specifically examined small PFS benefit with large OS benefit as well as small OS benefit with large PFS benefit, including differences in direction of PFS and OS outcomes. No evidence was identified that these are the result of poor study design, but may rather be due to the mechanism of action, specific disease, and population under study. Further work is needed to understand the mechanism of action of novel biologic/targeted agents to better understand their interaction with the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | - Pablo Lee
- Eli Lilly and Company, Indianapolis USA
| | | |
Collapse
|
227
|
|
228
|
Lin J, Shi J, Guo H, Yang X, Jiang Y, Long J, Bai Y, Wang D, Yang X, Wan X, Zhang L, Pan J, Hu K, Guan M, Huo L, Sang X, Wang K, Zhao H. Alterations in DNA Damage Repair Genes in Primary Liver Cancer. Clin Cancer Res 2019; 25:4701-4711. [PMID: 31068370 DOI: 10.1158/1078-0432.ccr-19-0127] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/16/2019] [Accepted: 05/03/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Alterations in DNA damage repair (DDR) genes produce therapeutic biomarkers. However, the characteristics and significance of DDR alterations remain undefined in primary liver cancer (PLC). EXPERIMENTAL DESIGN Patients diagnosed with PLC were enrolled in the trial (PTHBC, NCT02715089). Tumors and matched blood samples from participants were collected for a targeted next-generation sequencing assay containing exons of 450 cancer-related genes, including 31 DDR genes. The OncoKB knowledge database was used to identify and classify actionable alterations, and therapeutic regimens were determined after discussion by a multidisciplinary tumor board. RESULTS A total of 357 patients with PLC were enrolled, including 214 with hepatocellular carcinoma, 122 with ICC, and 21 with mixed hepatocellular-cholangiocarcinoma. A total of 92 (25.8%) patients had at least one DDR gene mutation, 15 of whom carried germline mutations. The most commonly altered DDR genes were ATM (5%) and BRCA1/2 (4.8%). The occurrence of DDR mutations was significantly correlated with a higher tumor mutation burden regardless of the PLC pathologic subtype. For DDR-mutated PLC, 26.1% (24/92) of patients possessed at least one actionable alteration, and the actionable frequency in DDR wild-type PLC was 18.9% (50/265). Eight patients with the BRCA mutation were treated by olaparib, and patients with BRCA2 germline truncation mutations showed an objective response. CONCLUSIONS The landscape of DDR mutations and their association with genetic and clinicopathologic features demonstrated that patients with PLC with altered DDR genes may be rational candidates for precision oncology treatment.
Collapse
Affiliation(s)
- Jianzhen Lin
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | | | | | - Xu Yang
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | | | - Junyu Long
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Yi Bai
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Dongxu Wang
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Xueshuai Wan
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Lei Zhang
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Jie Pan
- Department of Radiology, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Ke Hu
- Department of Radiotherapy, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Mei Guan
- Department of Medical Oncology, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Li Huo
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| | - Kai Wang
- OrigiMed, Shanghai, China.
- Zhejiang University International Hospital, Zhejiang, China
| | - Haitao Zhao
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China.
- Department of Liver Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
229
|
Tian Z, Gu Z, Wang X, Liu Z, Yao W, Wang J, Zhang P, Cai Q, Ge H. Efficacy and safety of apatinib in treatment of osteosarcoma after failed standard multimodal therapy: An observational study. Medicine (Baltimore) 2019; 98:e15650. [PMID: 31083265 PMCID: PMC6531132 DOI: 10.1097/md.0000000000015650] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recently, apatinib has been shown to be effective in treating sarcoma. This study aimed to assess the safety and efficacy of apatinib in the treatment of patients with osteosarcoma after failed of standard multimodal therapy and to compare the therapeutic effects of apatinib on osteosarcoma between high-dose group and low-dose group.A total of 27 patients with osteosarcoma who received apatinib between January 2016 and August 2017 were retrospectively reviewed. Among the 27 patients, the objective response rate (ORR) and the disease control rate (DCR) were 25.93% and 66.67%, respectively. The median of progression-free survival (m-PFS) was 3.5 months (95% confidence interval [CI], 2.5-4.8 months), and the median of overall survival (m-OS) was 9.5 months (95% CI, 7.8-10.5 months). There was no statistically significant difference in ORR (36.36% vs 18.75%), DCR (63.64% vs 68.75%), m-PFS (4.3 months [95% CI, 1.8-7 months) vs 3.35 months (95% CI, 1.8-4 months]), and m-OS (9.5 months [95% CI, 7.8-10.5 months] vs 9.4 months [95% CI, 7.8-10.8 months]) (P > .05) between the high-dose group (the average dose was 659 mg/qd) and the low-dose group (the average dose was 516 mg/qd). Most of the adverse events (AEs) were in grade 1 or grade 2. The main AEs in grade 3 were hypertension, rash, weight loss, hand-foot syndrome, and diarrhea.Apatinib is safe and effective in the treatment of advanced osteosarcoma. We recommend that the initial dose of apatinib should be 500 mg/qd in the treatment of osteosarcoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hong Ge
- Department of Radiation Oncology, The affiliated cancer hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
230
|
Persigehl T, Poeppel TD, Sedlaczek O. [Radiological response assessment of modern immunotherapy using iRECIST]. Radiologe 2019; 57:826-833. [PMID: 28812098 DOI: 10.1007/s00117-017-0289-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CLINICAL/METHODICAL ISSUE Modern immunotherapies in oncology show tumor response patterns differing from conventional chemotherapies including initial pseudo-progression. STANDARD RADIOLOGICAL METHODS Response evaluation criteria in solid tumors (RECIST 1.1) represent the currently most used response criteria for conventional chemotherapy of solid tumors. However, atypical response patterns of immunotherapies are not correctly classified using RECIST 1.1 so that the effectiveness is also incorrectly interpreted. METHODICAL INNOVATIONS In order to correctly interpret these atypical response patterns, special immune-related response criteria in solid tumors (iRECIST) have been published. In contrast to RECIST 1.1 according to iRECIST an initially unconfirmed progressive disease (iUPD) requires confirmation (iCPD) in clinically stable patients by subsequent control imaging after 4-8 weeks. New lesions are separately assessed within iRECIST. PERFORMANCE The iRECIST procedure allows a standardized objective assessment of a possible pseudo-progression which can occur in up to 10% of cases depending on the immunomodulating drug and tumor entity. ACHIEVEMENTS In principle, iRECIST was developed only for usage in trials testing modern immunotherapeutics. PRACTICAL RECOMMENDATIONS The iRECIST procedure might also be helpful as an additional objective response criterium for clinical treatment decisions, taking the limitations into account.
Collapse
Affiliation(s)
- T Persigehl
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - T D Poeppel
- Klinik für Nuklearmedizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122, Essen, Deutschland
| | - O Sedlaczek
- Abteilung Diagnostische und Interventionelle Radiologie, NCT Heidelberg (DKFZ & Universitätsklinikum HD), Im Neuenheimer Feld 460, 69120, Heidelberg, Deutschland
| |
Collapse
|
231
|
Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, Mak RH, Tamimi RM, Tempany CM, Swanton C, Hoffmann U, Schwartz LH, Gillies RJ, Huang RY, Aerts HJWL. Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin 2019; 69:127-157. [PMID: 30720861 PMCID: PMC6403009 DOI: 10.3322/caac.21552] [Citation(s) in RCA: 785] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Judgement, as one of the core tenets of medicine, relies upon the integration of multilayered data with nuanced decision making. Cancer offers a unique context for medical decisions given not only its variegated forms with evolution of disease but also the need to take into account the individual condition of patients, their ability to receive treatment, and their responses to treatment. Challenges remain in the accurate detection, characterization, and monitoring of cancers despite improved technologies. Radiographic assessment of disease most commonly relies upon visual evaluations, the interpretations of which may be augmented by advanced computational analyses. In particular, artificial intelligence (AI) promises to make great strides in the qualitative interpretation of cancer imaging by expert clinicians, including volumetric delineation of tumors over time, extrapolation of the tumor genotype and biological course from its radiographic phenotype, prediction of clinical outcome, and assessment of the impact of disease and treatment on adjacent organs. AI may automate processes in the initial interpretation of images and shift the clinical workflow of radiographic detection, management decisions on whether or not to administer an intervention, and subsequent observation to a yet to be envisioned paradigm. Here, the authors review the current state of AI as applied to medical imaging of cancer and describe advances in 4 tumor types (lung, brain, breast, and prostate) to illustrate how common clinical problems are being addressed. Although most studies evaluating AI applications in oncology to date have not been vigorously validated for reproducibility and generalizability, the results do highlight increasingly concerted efforts in pushing AI technology to clinical use and to impact future directions in cancer care.
Collapse
Affiliation(s)
- Wenya Linda Bi
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Ahmed Hosny
- Research Scientist, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Matthew B. Schabath
- Associate Member, Department of Cancer EpidemiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Maryellen L. Giger
- Professor of Radiology, Department of RadiologyUniversity of ChicagoChicagoIL
| | - Nicolai J. Birkbak
- Research Associate, The Francis Crick InstituteLondonUnited Kingdom
- Research Associate, University College London Cancer InstituteLondonUnited Kingdom
| | - Alireza Mehrtash
- Research Assistant, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Research Assistant, Department of Electrical and Computer EngineeringUniversity of British ColumbiaVancouverBCCanada
| | - Tavis Allison
- Research Assistant, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Research Assistant, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Omar Arnaout
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Christopher Abbosh
- Research Fellow, The Francis Crick InstituteLondonUnited Kingdom
- Research Fellow, University College London Cancer InstituteLondonUnited Kingdom
| | - Ian F. Dunn
- Associate Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Raymond H. Mak
- Associate Professor, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Rulla M. Tamimi
- Associate Professor, Department of MedicineBrigham and Women’s Hospital, Dana‐Farber Cancer Institute, Harvard Medical SchoolBostonMA
| | - Clare M. Tempany
- Professor of Radiology, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Charles Swanton
- Professor, The Francis Crick InstituteLondonUnited Kingdom
- Professor, University College London Cancer InstituteLondonUnited Kingdom
| | - Udo Hoffmann
- Professor of Radiology, Department of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Lawrence H. Schwartz
- Professor of Radiology, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Chair, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Robert J. Gillies
- Professor of Radiology, Department of Cancer PhysiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Raymond Y. Huang
- Assistant Professor, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Hugo J. W. L. Aerts
- Associate Professor, Departments of Radiation Oncology and Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Professor in AI in Medicine, Radiology and Nuclear Medicine, GROWMaastricht University Medical Centre (MUMC+)MaastrichtThe Netherlands
| |
Collapse
|
232
|
|
233
|
Alongi F, Fiorentino A, Gregucci F, Corradini S, Giaj-Levra N, Romano L, Rigo M, Ricchetti F, Beltramello A, Lunardi G, Mazzola R, Ruggieri R. First experience and clinical results using a new non-coplanar mono-isocenter technique (HyperArc™) for Linac-based VMAT radiosurgery in brain metastases. J Cancer Res Clin Oncol 2019; 145:193-200. [PMID: 30382369 DOI: 10.1007/s00432-018-2781-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 10/24/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Radiosurgery (SRS) or stereotactic fractionated radiotherapy (SFRT) is increasing in the treatment of brain metastases (BMs). Aim of the present study was to evaluate the safety and effectiveness of SRS/SFRT for BMs, using a new mono-isocenter non-coplanar solution (HyperArc™ Varian Medical System). METHODS BMs patients with a diameter inferior to 3 cm, a life expectancy of more than 3 months and a good performance status, were eligible for Linac-based volumetric modulated arc therapy (VMAT) SFRT/SRS with HyperArc™. A retrospective analysis of patients and BMs was performed. RESULTS From August 2017 to May 2018, 381 BMs in 64 patients were treated and 246 BMs (43 patients, median number of BMs: 5) of them were suitable for analysis. With a median FU time of 6 months, 244 out 246 (99%) BMs were controlled (18% complete response; 41% partial response, 40% stable disease), 2 BMs showed a progression, at the first control. No acute or late toxicities were reported. Median overall survival (OS) has not yet been achieved, while median time to progression was 5 months. In univariate analysis, statistically negative prognostic factors for OS were histology of primary tumor (p = 0.009): lung/breast cancer had better survival rates as compared to others. Cumulative intracranial volume disease ≥ 15 cc and systemic progression disease were independent prognostic factors for OS at univariate (p = 0.04; p = 0.005) and multivariate (p = 0.04; p = 0.009) analysis, respectively. CONCLUSION The present first clinical data show that SFRT/SRS with HyperArc™ is safe and effective for BMs patients. The utilization of SFRT/SRS for BMs is promising and should be further explored in randomized trials.
Collapse
Affiliation(s)
- Filippo Alongi
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
- University of Brescia, Piazza del Mercato 15, 25121, Brescia, Italy
| | - Alba Fiorentino
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Fabiana Gregucci
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy.
| | - Stefanie Corradini
- Department of Radiation Oncology, LMU University Hospital, Marchioninistr. 15, 81377, Munich, Germany
| | - Niccolo Giaj-Levra
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Luigi Romano
- Department of Radiology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Michele Rigo
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Francesco Ricchetti
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Alberto Beltramello
- Department of Radiology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Gianluigi Lunardi
- Department of Medical Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Rosario Mazzola
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| | - Ruggero Ruggieri
- Department of Radiation Oncology, IRCCS, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024, Negrar, Verona, Italy
| |
Collapse
|
234
|
Golan T, Milella M, Ackerstein A, Berger R. The changing face of clinical trials in the personalized medicine and immuno-oncology era: report from the international congress on clinical trials in Oncology & Hemato-Oncology (ICTO 2017). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:192. [PMID: 29282151 PMCID: PMC5745625 DOI: 10.1186/s13046-017-0668-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 01/10/2023]
Abstract
In the past decade, the oncology community has witnessed major advances in the understanding of cancer biology and major breakthroughs in several different therapeutic areas, from solid tumors to hematological malignancies; moreover, the advent of effective immunotherapy approaches, such as immune-checkpoint blockade, is revolutionizing treatment algorithms in almost all oncology disease areas. As knowledge evolves and new weapons emerge in the “war against cancer”, clinical and translational research need to adapt to a rapidly changing environment to effectively translate novel concepts into sustainable and accessible therapeutic options for cancer patients. With this in mind, translational cancer researchers, oncology professionals, treatment experts, CRO and industry leaders, as well as patient representatives gathered in London, 16-17 March 2017, for The International Congress on Clinical Trials in Oncology and Hemato-Oncology (ICTO2017), to discuss the changing face of oncology clinical trials in the new era of personalized medicine and immuno-oncology. A wide range of topics, including clinical trial design in immuno-oncology, biomarker-oriented drug development paths, statistical design and endpoint selection, challenges in the design and conduct of personalized medicine clinical trials, risk-based monitoring, financing and reimbursement, as well as best operational practices, were discussed in an open, highly interactive format, favoring networking among all relevant stakeholders. The most relevant data, approaches and issues emerged and discussed during the conference are summarized in this report.
Collapse
Affiliation(s)
- Talia Golan
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michele Milella
- Division of Medical Oncology 1, Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy.
| | - Aliza Ackerstein
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel
| | - Ranaan Berger
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
235
|
Lewis H, Ghasabeh M, Khoshpouri P, Kamel I, Pawlik T. Functional hepatic imaging as a biomarker of primary and secondary tumor response to loco-regional therapies. Surg Oncol 2017; 26:411-422. [DOI: 10.1016/j.suronc.2017.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
|
236
|
Goebel J, Hoischen J, Gramsch C, Schemuth HP, Hoffmann AC, Umutlu L, Nassenstein K. Tumor response assessment: comparison between unstructured free text reporting in routine clinical workflow and computer-aided evaluation based on RECIST 1.1 criteria. J Cancer Res Clin Oncol 2017; 143:2527-2533. [PMID: 28825135 DOI: 10.1007/s00432-017-2488-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Standardized computer-aided tumor response assessment is common in clinical trials. In contrast, unstructured free text reporting (UFTR) is common in daily routine. Therefore, this study aimed to discern and quantify differences between UFTR and computer-aided standardized tumor response evaluation based on RECIST 1.1 criteria (RECIST), serving as gold standard, in clinical workflow. METHODS One-hundred consecutive patients with cancer eligible for RECIST 1.1 evaluation, who received five follow-up CTs of the trunk, were retrospectively included. All UFTRs were assigned to RECIST response categories [complete response, partial response (PR), stable disease (SD), progressive disease (PD)]. All CTs were re-evaluated using dedicated software (mint lesion™) applying RECIST 1.1. The accordance in tumor response ratings was analyzed using Cohen's kappa. RESULTS At the first follow-up, 47 cases were rated differently with an SD underrepresentation and a PR and PD overrepresentation in UFTR. In the subsequent follow-ups, categorical differences were seen in 38, 44, 37, and 44%. Accordance between UFTR and RECIST was fair to moderate (Cohen's kappa: 0.356, 0.477, 0.390, 0.475, 0.376; always p < 0.001). Differences were mainly caused by the rating of even small tumor burden changes as PD or PR in UFTR or by comparison to the most recent prior CT scan in UFTR instead of comparison to nadir or baseline. CONCLUSIONS Significant differences in tumor response ratings were detected comparing UFTR and computer-aided standardized evaluation based on RECIST 1.1. Thus, standardized reporting should be implemented in daily routine workflow.
Collapse
Affiliation(s)
- Juliane Goebel
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| | - Julia Hoischen
- Department of Radiology, Evangelic Hospital Duesseldorf, Duesseldorf, Germany
| | - Carolin Gramsch
- Institute for Neuroradiology, University Hospital Giessen, Giessen, Germany
| | - Haemi P Schemuth
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Kai Nassenstein
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| |
Collapse
|
237
|
Perez-Lopez R, Roda D, Jimenez B, Brown J, Mateo J, Carreira S, Lopez J, Banerji U, Molife LR, Koh DM, Kaye SB, de Bono JS, Tunariu N, Yap TA. High frequency of radiological differential responses with poly(ADP-Ribose) polymerase (PARP) inhibitor therapy. Oncotarget 2017; 8:104430-104443. [PMID: 29262651 PMCID: PMC5732817 DOI: 10.18632/oncotarget.22303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/30/2017] [Indexed: 12/30/2022] Open
Abstract
Despite impressive clinical activity in patients with germline BRCA1 and BRCA2 (BRCA1/2) mutant cancers, antitumor responses to poly(ADP-Ribose) polymerase (PARP) inhibitors are variable. We set out to assess the rate of intrapatient radiological differential responses (RDR) to PARP inhibitors, its correlation with patient outcomes, and the identification of factors associated with RDR. We retrospectively reviewed all patients with advanced cancers from five early phase PARP inhibitor monotherapy trials. 113 patients (ovarian cancers 57.5%; breast cancers 23.9%) were included in this retrospective study; 46 (40.7%) patients developed RDR on PARP inhibitor monotherapy. We identified two patterns of RDR: early RDR (1st or 2nd on-treatment scans) in 69.6% of patients, and late RDR (penultimate or final scans) in 30.4% of patients. Early RDR was associated with shorter time to progression (TTP) (225 vs 367 days, HR:0.59, 95%CI 0.36-0.98; p=0.04) and overall survival (OS) (499 vs 857 days; HR:0.47, 95%CI 0.27-0.82, p=0.006). Seventy-nine (69.9%) patients had known germline BRCA1/2 mutations; 49.4% of these BRCA1/2 mutation carriers developed RDR versus 20.6% of patients with unknown or wildtype BRCA1/2 status. Harboring germline BRCA1/2 mutations was independently predictive for RDR (RR:2.93, 95% CI 1.08-7.90, p=0.03). Patients with germline BRCA1 mutations had worse TTP and OS than BRCA2 mutation carriers (212 vs 406 days, HR:0.58, 95% CI 0.36-0.94, p=0.023 and 515 vs 937 days; HR:0.49, 95% CI 0.29-0.83; p=0.007). RDR with PARP inhibitors are frequent, particularly in germline BRCA1/2 mutation carriers. These findings have clinical implications for patient outcomes and may reflect underlying intrapatient genomic heterogeneity.
Collapse
Affiliation(s)
- Raquel Perez-Lopez
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Desam Roda
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Begona Jimenez
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Jessica Brown
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Joaquin Mateo
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Juanita Lopez
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Udai Banerji
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - L. Rhoda Molife
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Dow-Mu Koh
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Stan B. Kaye
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Johann S. de Bono
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Nina Tunariu
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Timothy A. Yap
- The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
238
|
Morgan RL, Camidge DR. Reviewing RECIST in the Era of Prolonged and Targeted Therapy. J Thorac Oncol 2017; 13:154-164. [PMID: 29113950 DOI: 10.1016/j.jtho.2017.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 01/10/2023]
Abstract
Accurate assessment of disease response is the foundation of therapeutic trails, which is why the Response Evaluation Criteria in Solid Tumors (RECIST) serve as an international standard that investigators can utilize when examining patient outcomes. Nine years after the initial RECIST criteria were released, an update, RECIST 1.1, was published to improve on the initial criteria and address technologic advancements in imaging. Since then, advancements in both standard clinical and trial practices, combined with improvements in our understanding of cancer biology, have resulted in the identification of a number of limitations of the current RECIST 1.1, either in lack of clear guidance with regard to its best application or in potential benefit of capturing imaging-related data beyond standard categorical response details. As several of these situations reflect the consequences of prolonged control of metastatic disease by using targeted therapies, thoracic oncology has generated many of the key scenarios requiring elucidation and/or improvements. This article specifically examines current controversies in the interpretation and/or optimal utilization of RECIST 1.1, focusing on examples from thoracic oncology, and makes proposals, where possible, on how best to address these issues. These situations include addressing central nervous system versus extra-central nervous system response and progression, depth of response, oligoprogression versus polyprogression, continuation of systemic therapy after use of a local ablative therapy, and the impact of fluctuations in measurements bridging partial response and stable disease categories during prolonged therapy.
Collapse
Affiliation(s)
- Rustain L Morgan
- Department of Radiology, University of Colorado School of Medicine, Aurora, Colorado
| | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
239
|
RECIST response and variation of circulating tumour cells in phase 1 trials: A prospective multicentric study. Eur J Cancer 2017; 83:185-193. [DOI: 10.1016/j.ejca.2017.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/10/2017] [Indexed: 01/28/2023]
|
240
|
Lowery MA, Yu KH, Kelsen DP, Harding JJ, Bomalaski JS, Glassman DC, Covington CM, Brenner R, Hollywood E, Barba A, Johnston A, Liu KCW, Feng X, Capanu M, Abou-Alfa GK, O'Reilly EM. A phase 1/1B trial of ADI-PEG 20 plus nab-paclitaxel and gemcitabine in patients with advanced pancreatic adenocarcinoma. Cancer 2017; 123:4556-4565. [PMID: 28832976 DOI: 10.1002/cncr.30897] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/06/2017] [Accepted: 06/12/2017] [Indexed: 11/09/2022]
Abstract
BACKGROUND ADI-PEG 20 is a pegylated form of the arginine-depleting enzyme arginine deiminase. Normal cells synthesize arginine with the enzyme argininosuccinate synthetase (ASS1); ADI-PEG 20 selectively targets malignant cells, which lack ASS1. METHODS A single-arm, nonrandomized, open-label, phase 1/1B, standard 3 + 3 dose escalation with an expansion cohort of 9 patients at the recommended phase 2 dose (RP2D) was conducted. Patients who had metastatic pancreatic cancer, up to 1 line of prior treatment (the dose-escalation cohort) or no prior treatment (the expansion cohort), and an Eastern Cooperative Oncology Group performance status of 0 to 1 were included. Patients received both gemcitabine (1000 mg/m2 ) and nab-paclitaxel (125 mg/m2 ) for 3 of 4 weeks and intramuscular ADI-PEG 20 at 18 mg/m2 weekly (cohort 1) or at 36 mg/m2 weekly (cohort 2 and the expansion cohort).The primary endpoint was to determine the maximum tolerated dose and RP2D of ADI-PEG 20 in combination with nab-paclitaxel and gemcitabine. RESULTS Eighteen patients were enrolled. No dose-limiting toxicities (DLTs) were observed in cohort 1; cohort 2 was expanded to 6 patients because of 1 DLT occurrence (a grade 3 elevation in bilirubin, aspartate aminotransferase, and alanine aminotransferase). The most frequent adverse events (AEs) of any grade were neutropenia, thrombocytopenia, leukopenia, anemia, peripheral neuropathy, and fatigue; all 18 patients experienced grade 3/4 AEs. The most frequent grade 3/4 toxicities, regardless of the relation with any drugs, included neutropenia (12 patients or 67%), leukopenia (10 patients or 56%), anemia (8 patients or 44%), and lymphopenia (6 patients or 33%). The RP2D for ADI-PEG 20 was 36 mg/m2 weekly in combination with standard-dose gemcitabine and nab-paclitaxel. The overall response rate among patients treated at the RP2D in the first-line setting was 45.5% (5 of 11).The median progression-free survival time for these patients treated at the RP2D was 6.1 months (95% confidence interval, 5.3-11.2 months), and the median overall survival time was 11.3 months (95% confidence interval, 6.7 months to not reached). CONCLUSIONS ADI-PEG 20 was well tolerated in combination with gemcitabine and nab-paclitaxel. Activity was observed in previously treated and untreated patients with advanced pancreatic cancer and in patients with ASS1-deficient and -proficient tumors. Cancer 2017;123:4556-4565. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Maeve A Lowery
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - David Paul Kelsen
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - James J Harding
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | | | | | | | - Robin Brenner
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | | | - Marinela Capanu
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
241
|
de Bree R, Wolf GT, de Keizer B, Nixon IJ, Hartl DM, Forastiere AA, Haigentz M, Rinaldo A, Rodrigo JP, Saba NF, Suárez C, Vermorken JB, Ferlito A. Response assessment after induction chemotherapy for head and neck squamous cell carcinoma: From physical examination to modern imaging techniques and beyond. Head Neck 2017; 39:2329-2349. [PMID: 28815841 PMCID: PMC5656833 DOI: 10.1002/hed.24883] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/27/2017] [Accepted: 05/31/2017] [Indexed: 01/27/2023] Open
Abstract
Significant correlations between the response to induction chemotherapy and success of subsequent radiotherapy have been reported and suggest that the response to induction chemotherapy is able to predict a response to radiotherapy. Therefore, induction chemotherapy may be used to tailor the treatment plan to the individual patient with head and neck cancer: following the planned subsequent (chemo)radiation schedule, planning a radiation dose boost, or reassessing the modality of treatment (eg, upfront surgery). Findings from reported trials suggest room for improvement in clinical response assessment after induction chemotherapy, but an optimal method has yet to be identified. Historically, indices of treatment efficacy in solid tumors have been based solely on systematic assessment of tumor size. However, functional imaging (eg, fluorodeoxyglucose‐positron emission tomography (FDG‐PET) potentially provides an earlier indication of response to treatment than conventional imaging techniques. More advanced imaging techniques are still in an exploratory phase and are not ready for use in clinical practice.
Collapse
Affiliation(s)
- Remco de Bree
- Department of Head and Neck Surgical Oncology, UMC Utrecht Cancer Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gregory T Wolf
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Bart de Keizer
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Iain J Nixon
- Ear, Nose, and Throat Department, NHS Lothian, Edinburgh, UK
| | - Dana M Hartl
- Department of Otolaryngology - Head and Neck Surgery, Institut Gustave Roussy, Villejuif Cedex, France.,Laboratoire de Phonétique et de Phonologie, Sorbonne Nouvelle, Paris, France
| | - Arlene A Forastiere
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Missak Haigentz
- Department of Medicine, Division of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Juan P Rodrigo
- Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Spain.,Department of Otolaryngology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, The Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Carlos Suárez
- Department of Otolaryngology, Hospital Universitario Central de Asturias, Oviedo, Spain.,Fundación de Investigación e Innovación Biosanitaria del Principado de Asturias, Oviedo, Spain
| | - Jan B Vermorken
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Alfio Ferlito
- Coordinator of the International Head and Neck Scientific Group
| |
Collapse
|
242
|
Dercle L, Ammari S, Bateson M, Durand PB, Haspinger E, Massard C, Jaudet C, Varga A, Deutsch E, Soria JC, Ferté C. Limits of radiomic-based entropy as a surrogate of tumor heterogeneity: ROI-area, acquisition protocol and tissue site exert substantial influence. Sci Rep 2017; 7:7952. [PMID: 28801575 PMCID: PMC5554130 DOI: 10.1038/s41598-017-08310-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 07/10/2017] [Indexed: 01/19/2023] Open
Abstract
Entropy is a promising quantitative imaging biomarker for characterizing cancer imaging phenotype. Entropy has been associated with tumor gene expression, tumor metabolism, tumor stage, patient prognosis, and treatment response. Our hypothesis states that tumor-specific biomarkers such as entropy should be correlated between synchronous metastases. Therefore, a significant proportion of the variance of entropy should be attributed to the malignant process. We analyzed 112 patients with matched/paired synchronous metastases (SM#1 and SM#2) prospectively enrolled in the MOSCATO-01 clinical trial. Imaging features were extracted from Regions Of Interest (ROI) delineated on CT-scan using TexRAD software. We showed that synchronous metastasis entropy was correlated across 5 Spatial Scale Filters: Spearman's Rho ranged between 0.41 and 0.59 (P = 0.0001, Bonferroni correction). Multivariate linear analysis revealed that entropy in SM#1 is significantly associated with (i) primary tumor type; (ii) entropy in SM#2 (same malignant process); (iii) ROI area size; (iv) metastasis site; and (v) entropy in the psoas muscle (reference tissue). Entropy was a logarithmic function of ROI area in normal control tissues (aorta, psoas) and in mathematical models (P < 0.01). We concluded that entropy is a tumor-specific metric only if confounding factors are corrected.
Collapse
Affiliation(s)
- Laurent Dercle
- INSERM U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.
- Département de l'imagerie médicale, Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France.
- Department of Radiology, Columbia University Medical Center, New York, New York, USA.
| | - Samy Ammari
- Département de l'imagerie médicale, Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
| | | | - Paul Blanc Durand
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
| | - Eva Haspinger
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
| | - Cyril Jaudet
- Department of Radiotherapy, UZ Brussel, Brussels, Belgium
| | - Andrea Varga
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
| | - Eric Deutsch
- Département de radiothérapie, Gustave Roussy Cancer Campus, Université Paris Saclay, F-94805, Villejuif, France
- INSERM U981, Biomarqueurs prédictifs et nouvelles stratégies en oncologie, Université Paris Sud, Gustave Roussy, Villejuif, France
| | - Jean-Charles Soria
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France
- INSERM U981, Biomarqueurs prédictifs et nouvelles stratégies en oncologie, Université Paris Sud, Gustave Roussy, Villejuif, France
- INSERM U1030, Paris Sud University, Gustave Roussy, Villejuif, France
| | - Charles Ferté
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, F-94805, Villejuif, France.
- INSERM U981, Biomarqueurs prédictifs et nouvelles stratégies en oncologie, Université Paris Sud, Gustave Roussy, Villejuif, France.
- INSERM U1030, Paris Sud University, Gustave Roussy, Villejuif, France.
| |
Collapse
|
243
|
Dubreuil J, Cachin F, Berriolo-Ridinger A, Skanjeti A. Critères d’interprétation en imagerie cancérologique solide : RECIST, PERCIST…. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2017. [DOI: 10.1016/j.mednuc.2017.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
244
|
Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, Lin NU, Litière S, Dancey J, Chen A, Hodi FS, Therasse P, Hoekstra OS, Shankar LK, Wolchok JD, Ballinger M, Caramella C, de Vries EGE. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017; 18:e143-e152. [PMID: 28271869 PMCID: PMC5648544 DOI: 10.1016/s1470-2045(17)30074-8] [Citation(s) in RCA: 1673] [Impact Index Per Article: 209.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/25/2016] [Accepted: 11/30/2016] [Indexed: 12/11/2022]
Abstract
Tumours respond differently to immunotherapies compared with chemotherapeutic drugs, raising questions about the assessment of changes in tumour burden-a mainstay of evaluation of cancer therapeutics that provides key information about objective response and disease progression. A consensus guideline-iRECIST-was developed by the RECIST working group for the use of modified Response Evaluation Criteria in Solid Tumours (RECIST version 1.1) in cancer immunotherapy trials, to ensure consistent design and data collection, facilitate the ongoing collection of trial data, and ultimate validation of the guideline. This guideline describes a standard approach to solid tumour measurements and definitions for objective change in tumour size for use in trials in which an immunotherapy is used. Additionally, it defines the minimum datapoints required from future trials and those currently in development to facilitate the compilation of a data warehouse to use to later validate iRECIST. An unprecedented number of trials have been done, initiated, or are planned to test new immune modulators for cancer therapy using a variety of modified response criteria. This guideline will allow consistent conduct, interpretation, and analysis of trials of immunotherapies.
Collapse
Affiliation(s)
- Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada.
| | | | | | - Robert Ford
- Clinical Trials Imaging Consulting, LLC, Belle Mead, NJ, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA
| | - Sumithra Mandrekar
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Janet Dancey
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Alice Chen
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Lalitha K Shankar
- Diagnostic Imaging Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcus Ballinger
- Weill Cornell Medical and Graduate Colleges, New York, NY, USA; Ludwig Institute for Cancer Research, New York, NY, USA; Genentech Inc, San Francisco, CA, USA
| | | | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
245
|
|