101
|
Damato A, Bergamo F, Antonuzzo L, Nasti G, Iachetta F, Romagnani A, Gervasi E, Larocca M, Pinto C. FOLFOXIRI/Bevacizumab Plus Nivolumab as First-Line Treatment in Metastatic Colorectal Cancer RAS/BRAF Mutated: Safety Run-In of Phase II NIVACOR Trial. Front Oncol 2021; 11:766500. [PMID: 34970487 PMCID: PMC8712943 DOI: 10.3389/fonc.2021.766500] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
The NIVACOR trial is a phase II study assessing the efficacy and safety of nivolumab in combination with FOLFOXIRI/bevacizumab in first-line setting in patients affected by metastatic colorectal cancer (mCRC) RAS/BRAF mutated. We report safety run-in results in the first 10 patients enrolled. Patients received triplet chemotherapy with FOLFOXIRI scheme plus bevacizumab, in association with nivolumab every 2 weeks for 8 cycles (induction phase) followed by bevacizumab plus nivolumab every 2 weeks (maintenance phase), until progression of disease or unacceptable toxicities. The first ten patients were evaluated: 7 experienced at least one adverse event (AE) related to FOLFOXIRI/bevacizumab and 2 related to nivolumab. The most frequent grade 1-2 AEs related to FOLFOXIRI/bevacizumab were diarrhea and fatigue (71%), nausea and vomiting (57%); 3 (43%) had grade 3-4 neutropenia, and 2 (20%) patients developed grade 1-2 AEs nivolumab related: skin rash and salivary gland infection. Two patients delayed the dose because of serious AEs, proteinuria and salivary gland infection; one patient discontinued experimental treatment due to the ileo-urethral fistula and concurrent Clostridium infection diarrhea. No treatment- related death occurred. The safety run-in analysis of NIVACOR trial reassured using co-administration of FOLFOXIRI/bevacizumab and nivolumab was well tolerated with an acceptable toxicity profile. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/, (NCT04072198).
Collapse
Affiliation(s)
- Angela Damato
- Medical Oncology Unit, Azienda USL (Unità Sanitaria Locale)- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) di Reggio Emilia, Reggio Emilia, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Bergamo
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology Istituto Veneto Oncologico (IOV) - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Guglielmo Nasti
- Medical Oncology, Abdominal Department, National Cancer Institute G. Pascale Foundation, Naples, Italy
| | - Francesco Iachetta
- Medical Oncology Unit, Azienda USL (Unità Sanitaria Locale)- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandra Romagnani
- Medical Oncology Unit, Azienda USL (Unità Sanitaria Locale)- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) di Reggio Emilia, Reggio Emilia, Italy
| | - Erika Gervasi
- Medical Oncology Unit, Azienda USL (Unità Sanitaria Locale)- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) di Reggio Emilia, Reggio Emilia, Italy
| | - Mario Larocca
- Medical Oncology Unit, Azienda USL (Unità Sanitaria Locale)- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) di Reggio Emilia, Reggio Emilia, Italy
| | - Carmine Pinto
- Medical Oncology Unit, Azienda USL (Unità Sanitaria Locale)- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
102
|
Gao L, Xue J, Liu X, Cao L, Wang R, Lei L. A risk model based on autophagy-related lncRNAs for predicting prognosis and efficacy of immunotherapy and chemotherapy in gastric cancer patients. Aging (Albany NY) 2021; 13:25453-25465. [PMID: 34897033 PMCID: PMC8714132 DOI: 10.18632/aging.203765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022]
Abstract
Long non-coding RNAs (lncRNAs) are a class of non-protein-coding RNAs essential to the occurrence and development of gastric cancer (GC). We aimed to identify critical lncRNA pairs to construct a prognostic model and assess its performances in prognosis and efficacy prediction in GC patients receiving immunotherapy and chemotherapy. We searched transcriptome and clinical data of GC patients from The Cancer Genome Atlas (TCGA) database. Autophagy-related lncRNAs were identified using co-expression network analysis, and lncRNA pairs with prognostic value were selected using pairwise transcriptome analysis. The gene pairs were subjected to LASSO algorithm for identification of optimal gene pairs for risk model construction. Patients were classified into the low-risk and high-risk groups with the RiskScore as a cutoff. Finally, 9 optimal gene pairs were identified in the LASSO algorithm model for construction of a lncRNA prognostic risk model. For predictive performances, it successfully predicted a shorter survival of high-risk patients than that obtained in low-risk individuals (P < 0.001). It showed moderate AUC (area under the curve) values for 1-, 2-, and 3-year overall survival prediction of 0.713 and could serve as an independent predictor for GC prognosis. Compared to the low-risk group, high-risk patients had higher expressions of marker genes for immune checkpoint inhibitors (ICIs) and showed higher sensitivity to the chemotherapy agents, rapamycin, bexarotene, and bicalutamide. Our findings demonstrate a robust prognostic model based on nine autophagy-related lncRNA pairs for GC. It acts as an independent predictor for survival and efficacy prediction of immunotherapy and chemotherapy in GC patients.
Collapse
Affiliation(s)
- Lei Gao
- Department of Gastroenterology, The First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Juan Xue
- Department of Clinical Laboratory, The First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiaomin Liu
- Department of Gastroenterology, The First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Lei Cao
- Department of Gastroenterology, The First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ruifang Wang
- Department of Gastroenterology, The First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Liangliang Lei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
103
|
Kamposioras K, Ntellas P, Nikolaou M, Germetaki T, Gazouli I, Dadouli K, Zarkavelis G, Amylidi AL, Tolia M, Mauri D. Immunotherapy Efficacy in the Initial Lines of Treatment in Advanced Upper Gastrointestinal Malignancies: A Systematic Review of the Literature. JNCI Cancer Spectr 2021; 5:pkab088. [PMID: 34926989 PMCID: PMC8677514 DOI: 10.1093/jncics/pkab088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/11/2021] [Accepted: 10/07/2021] [Indexed: 12/30/2022] Open
Abstract
Background The therapeutic role of immune checkpoint inhibitors (ICIs) has represented the cutting edge of clinical research in upper gastrointestinal (GI) malignancies, with these agents now included in the armamentarium of treatment options for advanced gastric and esophageal cancers. Methods We performed a systematic literature review and pooled analysis to map out the currently available robust clinical evidence for the use of ICIs in upper GI cancers. Immunotherapy (IO), either as monotherapy or in combination with chemotherapy, and its role in first-line, maintenance, and second-line settings, as well as in specific clinical and biological subgroups, were critically appraised. All statistical tests were 2-sided. Results ICIs, in combination with chemotherapy, have provided statistically significant overall survival benefit in the first-line setting in gastric and gastro-esophageal adenocarcinomas (hazard ratio [HR] = 0.83, 95% confidence interval [CI] = 0.76 to 0.90, P < .001; based on 4 studies) and esophageal squamous cell carcinoma (HR = 0.72, 95% CI = 0.64 to 0.81, P < .001; based on 3 studies), albeit with heterogeneous efficacy according to biomarker expression. Patients with esophageal squamous cell carcinoma, and in particular high programmed cell death ligand-1 expression, derive survival benefit when treated with IO in the second-line setting (HR = 0.74, 95% CI = 0.68 to 0.82, P < .001; for any level of programmed cell death ligand-1 expression). Clinical trials interrogating the combination of IO with chemotherapy in second-line treatment should be seriously considered in upper GI adenocarcinomas. The role of maintenance IO after initial disease control is still unclear and cannot be recommended. Impressive response rates and survival benefit from IO have been reported in patients with microsatellite instability-high tumors (HR = 0.33, 95% CI = 0.19 to 0.57, P < .001), and this warrants further prospective biomarker-driven studies. Conclusions IO is changing the treatment landscape in upper GI malignancies. The rapidly developing evidence in the field needs to be critically appraised while further validation of the existing information from ongoing trials is awaited.
Collapse
Affiliation(s)
| | - Panagiotis Ntellas
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Michail Nikolaou
- Department of Medical Oncology, Agios Savvas Anticancer Hospital, Athens, Greece
| | - Theodora Germetaki
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Ioanna Gazouli
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Katerina Dadouli
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Anna-Lea Amylidi
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Maria Tolia
- Department of Radiotherapy, School of Medicine, University of Crete, Heraklion, Greece
| | - Davide Mauri
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
104
|
Soeratram TTD, Creemers A, Meijer SL, de Boer OJ, Vos W, Hooijer GKJ, van Berge Henegouwen MI, Hulshof MCCM, Bergman JJGHM, Lei M, Bijlsma MF, Ylstra B, van Grieken NCT, van Laarhoven HWM. Tumor-immune landscape patterns before and after chemoradiation in resectable esophageal adenocarcinomas. J Pathol 2021; 256:282-296. [PMID: 34743329 PMCID: PMC9299918 DOI: 10.1002/path.5832] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/27/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapy is a new anti‐cancer treatment option, showing promising results in clinical trials. To investigate potential immune biomarkers in esophageal adenocarcinoma (EAC), we explored immune landscape patterns in the tumor microenvironment before and after neoadjuvant chemoradiation (nCRT). Sections from matched pretreatment biopsies and post‐nCRT resection specimens (n = 188) were stained for (1) programmed death‐ligand 1 (PD‐L1, CD274); (2) programmed cell death protein 1 (PD‐1, CD279), forkhead box P3 (FOXP3), CD8, pan‐cytokeratin multiplex; and (3) an MHC class I, II duplex. The densities of tumor‐associated immune cells (TAICs) were calculated using digital image analyses and correlated to histopathological nCRT response [tumor regression grade (TRG)], survival, and post‐nCRT immune patterns. PD‐L1 positivity defined by a combined positive score of >1 was associated with a better response post‐nCRT (TRG 1–3 versus 4, 5, p = 0.010). In addition, high combined mean densities of CD8+, FOXP3+, and PD‐1+ TAICs in the tumor epithelium and stroma of biopsies were associated with a better response (TRG 1–3 versus 4, 5, p = 0.025 and p = 0.044, respectively). Heterogeneous TAIC density patterns were observed post‐nCRT, with significantly higher CD8+ and PD‐1+ TAIC mean densities compared with biopsies (both p = 0.000). Three immune landscape patterns were defined post‐nCRT: ‘inflamed’, ‘invasive margin’, and ‘desert’, of which ‘inflamed’ was the most frequent (57%). Compared with matched biopsies, resection specimens with ‘inflamed’ tumors showed a significantly higher increase in CD8+ density compared with non‐inflamed tumors post‐nCRT (p = 0.000). In this cohort of EAC patients, higher TAIC densities in pretreatment biopsies were associated with response to nCRT. This warrants future research into the potential of the tumor‐immune landscape for patient stratification and novel (immune) therapeutic strategies. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tanya T D Soeratram
- Department of Pathology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Aafke Creemers
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sybren L Meijer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Onno J de Boer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Wim Vos
- Department of Pathology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerrit K J Hooijer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Mark I van Berge Henegouwen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Maarten C C M Hulshof
- Department of Radiotherapy, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jacques J G H M Bergman
- Department of Gastroenterology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Ming Lei
- Bristol-Myers Squibb, Princeton, NJ, USA
| | - Maarten F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Bauke Ylstra
- Department of Pathology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Nicole C T van Grieken
- Department of Pathology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
105
|
Jian D, Qian C, Wang D, Ma Q, Wang L, Li C, Xu M, Dai N, Chen Q, He J, Zhang H, Yuan M, Chen R, Chao R, Feng Y. Conversion therapy with tislelizumab for high microsatellite instability, unresectable stage III gastric cancer: a case report. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1489. [PMID: 34734041 PMCID: PMC8506721 DOI: 10.21037/atm-21-4295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/18/2021] [Indexed: 12/20/2022]
Abstract
Gastric cancer (GC) is the fifth-highest ranked cancer for incidence and second for mortality from cancer worldwide. Conversion therapy has recently emerged as an alternative therapy for advanced/metastatic GC patients who are unable to undergo surgical resection at the time of diagnosis. Herein, we present the case of a patient with unresectable stage III GC of high microsatellite instability (MSI), high tumor mutation burden (TMB), and Epstein-Barr virus (EBV) positive. The patient received conversion therapy involving a combination of chemotherapy and immunotherapy regimens. After 3 courses of chemotherapy combined with tislelizumab, the patient underwent laparoscopic radical total gastrectomy. The pathological examination demonstrated that there was no cancerous tissue at the proximal or distal end of the tumor and no lymph node metastases in the lesser or greater curvature, indicating a pathologic complete response. Thereafter, the patient continued tislelizumab treatment to prevent postoperative carcinoma recurrence and metastasis, and to improve prognosis. In conclusion, our study confirmed that chemotherapy combined with immunotherapy is a promising conversion therapy for GC patients with locally unresectable lesions or distant lymph node metastasis, and these findings warrant large-scale clinical studies. This report highlights the clinical importance of next-generation sequencing technology in investigating therapeutic strategy to provide the maximal clinical benefit for patients with GC.
Collapse
Affiliation(s)
- Dan Jian
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Chengyuan Qian
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Dong Wang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiang Ma
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, China
| | - Li Wang
- Department of Gastric & Colorectal Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Chunxue Li
- Department of Gastric & Colorectal Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingfang Xu
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Dai
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Juan He
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | | | | | | | - Rui Chao
- Department of Orthopaedic Surgery, Chongqing Emergency Medical Center, The Fourth People's Hospital of Chongqing, Chongqing University Central Hospital, Chongqing, China
| | - Yan Feng
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
106
|
de la Fouchardiere C, Decoster L, Samalin E, Terret C, Kenis C, Droz JP, Coutzac C, Smyth E. Advanced oesophago-gastric adenocarcinoma in older patients in the era of immunotherapy. A review of the literature. Cancer Treat Rev 2021; 100:102289. [PMID: 34583303 DOI: 10.1016/j.ctrv.2021.102289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022]
Abstract
Gastric (G) and gastro-esophageal junction (GEJ) adenocarcinomas are of the most common and deadly cancers worldwide and affect mainly patients over 70 years at diagnosis. Older age has been associated in gastric cancers with distal tumour location, well-differentiated adenocarcinoma and microsatellite instability and is not identified itself as an independent prognostic factor. As immune checkpoint inhibitors recently changed the landmark of advanced G and GEJ adenocarcinomas treatment, we decided to perform a literature review to define the evidence-level of clinical data in older patients. This work underlined the lasting low -inclusion rate of older patients and -implementation rate of frailty screening tools in clinical trials in G/GEJ carcinomas. In the first-line metastatic setting, two prospective randomized phase III studies have specifically assessed the efficacy of chemotherapy in older patients with HER2-negative gastric cancers, demonstrating the feasibility of reduced dose oxaliplatin-based chemotherapy regimen in this population. Only few data are available in HER2-positive tumors, or in the second-line setting. Furthermore, no specific trial with immune checkpoint inhibitors was performed in older frail patients whereas their benefit/adverse events ratio make them attractive candidates in this patient's population. We conclude that older fit patients can be treated in the same way as younger ones and included in clinical trials. Improving the outcome of older frail patients should be the oncology community next focus by implementing targeted interventions before initiating cancer therapy and designing specific clinical trials. Frailty screening tools and geriatric data collection have to be implemented in routine-practice and clinical trials.
Collapse
Affiliation(s)
| | - L Decoster
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | - E Samalin
- Medical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ. Montpellier, Montpellier, France.
| | - C Terret
- Medical Oncology Department, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - C Kenis
- Department of General Medical Oncology and Geriatric Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Public Health and Primary Care, Academic Centre for Nursing and Midwifery, KU Leuven - University of Leuven, Leuven, Belgium.
| | - J P Droz
- Medical Oncology, Claude-Bernard Lyon1 University, Lyon, France.
| | - C Coutzac
- Medical Oncology Department, Centre Léon Bérard, 28 rue Laennec, Lyon, France.
| | - E Smyth
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| |
Collapse
|
107
|
Co-Expression with Membrane CMTM6/4 on Tumor Epithelium Enhances the Prediction Value of PD-L1 on Anti-PD-1/L1 Therapeutic Efficacy in Gastric Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13205175. [PMID: 34680324 PMCID: PMC8533876 DOI: 10.3390/cancers13205175] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Immunotherapeutic efficacy is low even in PD-L1 positive patients with advanced gastric adenocarcinoma. Based on the results of 6-color multiplex immunofluorescence staining of the gastric tumor tissues in tissue array and 48-case pre-immunotherapy patients, a better prognostic value was found in the membrane co-expression of CMTM6/4 and PD-L1 in tumor epithelial cells than PD-L1 alone. The membrane co-expression of CMTM6/4 and PD-L1 can be used as a valuable tool for precision pre-immunotherapy patient screening in gastric adenocarcinoma. Abstract Anti-PD-1/L1 immunotherapy has been intensively used in heavily treated population with advanced gastric adenocarcinoma. However, the immunotherapeutic efficacy is low even in PD-L1 positive patients. We aimed to establish a new strategy based on the co-expression of CMTM6/4 and PD-L1 for patient stratification before immunotherapy. By analyzing the data obtained from TCGA and single-cell RNA sequencing at the mRNA level, and 6-color multiplex immunofluorescence staining of tumor tissues in tissue array and 48-case pre-immunotherapy patients at the protein level, we found that CMTM6/4 and PD-L1 co-expressed in both epithelial and mesenchymal regions of gastric adenocarcinoma. The tumor tissues had higher levels of CMTM6/4 expression than their adjacent ones. A positive correlation was found between the expression of CMTM6/4 and the expression of PD-L1 in tumor epithelium. Epithelial co-expression of CMTM6/4 and PD-L1 in gastric tumor region was associated with shorter overall survival but better short-term response to anti-PD-1/L1 immunotherapy. Thus, we developed a predictive model and three pathological patterns based on the membrane co-expression of CMTM6/4 and PD-L1 in tumor epithelial cells for pre-immunotherapy patient screening in gastric adenocarcinoma.
Collapse
|
108
|
Ruiz Hispán E, Pedregal M, Cristobal I, García-Foncillas J, Caramés C. Immunotherapy for Peritoneal Metastases from Gastric Cancer: Rationale, Current Practice and Ongoing Trials. J Clin Med 2021; 10:4649. [PMID: 34682772 PMCID: PMC8539177 DOI: 10.3390/jcm10204649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
Peritoneal metastases from gastric cancer play a key role in the fatal prognosis of the disease. The lack of efficacy of actual therapeutic approaches together with the outcomes achieved with checkpoint inhibitors in gastric cancer compel us to address the current state-of-the-art immunotherapy treatment of peritoneal dissemination. The immunogenicity of the peritoneum has been described to be particularly active at omentum and peritoneal lymph nodes. Also, both innate and acquired immunity seems to be involved at different molecular levels. Recent works show PDL1 expression being less present at the peritoneal level; however, some clinical trials have begun to yield results. For example, the ATTRACTION-2 trial has demonstrated the activity of Nivolumab in heavily pretreated patients even though peritoneal metastases were diagnosed in a 30% of them. Despite positive results in the metastatic setting, peritoneal responses to systemic checkpoint inhibitors remains unclear, therefore, new strategies for intraperitoneal immunotherapy are being proposed for different ongoing clinical trials.
Collapse
Affiliation(s)
- Eva Ruiz Hispán
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Manuel Pedregal
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Ion Cristobal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM Madrid, 28040 Madrid, Spain;
| | - Jesús García-Foncillas
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Cristina Caramés
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| |
Collapse
|
109
|
Yang Y, Zhang Z, Chen Q, You Y, Li X, Chen T. Functionalized Selenium Nanoparticles Synergizes With Metformin to Treat Breast Cancer Cells Through Regulation of Selenoproteins. Front Bioeng Biotechnol 2021; 9:758482. [PMID: 34708029 PMCID: PMC8543061 DOI: 10.3389/fbioe.2021.758482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 08/26/2021] [Indexed: 12/19/2022] Open
Abstract
Owing to high blood sugar level and chronic inflammation, diabetes tend to cause the overproduction of free radicals in body, which will damage tissue and cells, reduce autoimmunity, and greatly increase the incidence of tumors. Selenium nanoparticles (SeNPs) exhibit high antioxidant activity with anti-tumor ability. In addition, metformin is considered as a clinical drug commonly for the treatment of stage II diabetes. Therefore, in this study, different functionalized SeNPs combined with metformin were performed to detect the feasibility for cancer therapy. The combination of Tween 80 (TW80)-SeNPs and metformin was found to have a synergistic effect on MCF-7 cells. The mechanism of this synergistic effect involved in the induction of DNA damage by affecting the generation of reactive oxygen species through selenoproteins; the upregulation of DNA-damage-related proteins including p-ATM, p-ATR, and p38; the promotion of p21 expression; and the downregulation of cyclin-dependent kinases and cyclin-related proteins causing cell cycle arrest. Furthermore, the expression of AMPK was affected, which in turn to regulate the mitochondrial membrane potential to achieve the synergistic treatment effect.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoling Li
- Department of Chemistry, and Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| | - Tianfeng Chen
- Department of Chemistry, and Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| |
Collapse
|
110
|
Klein S, Duda DG. Machine Learning for Future Subtyping of the Tumor Microenvironment of Gastro-Esophageal Adenocarcinomas. Cancers (Basel) 2021; 13:4919. [PMID: 34638408 PMCID: PMC8507866 DOI: 10.3390/cancers13194919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor progression involves an intricate interplay between malignant cells and their surrounding tumor microenvironment (TME) at specific sites. The TME is dynamic and is composed of stromal, parenchymal, and immune cells, which mediate cancer progression and therapy resistance. Evidence from preclinical and clinical studies revealed that TME targeting and reprogramming can be a promising approach to achieve anti-tumor effects in several cancers, including in GEA. Thus, it is of great interest to use modern technology to understand the relevant components of programming the TME. Here, we discuss the approach of machine learning, which recently gained increasing interest recently because of its ability to measure tumor parameters at the cellular level, reveal global features of relevance, and generate prognostic models. In this review, we discuss the relevant stromal composition of the TME in GEAs and discuss how they could be integrated. We also review the current progress in the application of machine learning in different medical disciplines that are relevant for the management and study of GEA.
Collapse
Affiliation(s)
- Sebastian Klein
- Gerhard-Domagk-Institute for Pathology, University Hospital Münster, 48149 Münster, Germany
- Institute for Pathology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Dan G. Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02478, USA
| |
Collapse
|
111
|
Zhang SX, Liu W, Ai B, Sun LL, Chen ZS, Lin LZ. Current Advances and Outlook in Gastric Cancer Chemoresistance: A Review. Recent Pat Anticancer Drug Discov 2021; 17:26-41. [PMID: 34587888 DOI: 10.2174/1574892816666210929165729] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/19/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Surgical resection of the lesion is the standard primary treatment of gastric cancer. Unfortunately, most patients are already in the advanced stage of the disease when they are diagnosed with gastric cancer. Alternative therapies, such as radiation therapy and chemotherapy, can achieve only very limited benefits. The emergence of cancer drug resistance has always been the major obstacle to the cure of tumors. The main goal of modern cancer pharmacology is to determine the underlying mechanism of anticancer drugs. OBJECTIVE Here, we mainly review the latest research results related to the mechanism of chemotherapy resistance in gastric cancer, the application of natural products in overcoming the chemotherapy resistance of gastric cancer, and the new strategies currently being developed to treat tumors based on immunotherapy and gene therapy. CONCLUSION The emergence of cancer drug resistance is the main obstacle in achieving alleviation and final cure for gastric cancer. Mixed therapies are considered to be a possible way to overcome chemoresistance. Natural products are the main resource for discovering new drugs specific for treating chemoresistance, and further research is needed to clarify the mechanism of natural product activity in patients. .
Collapse
Affiliation(s)
- Sheng-Xiong Zhang
- Guangdong Province Work Injury Rehabilitation Hospital, Guangzhou, 510440. China
| | - Wei Liu
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006. China
| | - Bo Ai
- Huazhong University of Science and Technology, Wuhan, 430030. China
| | - Ling-Ling Sun
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405. China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, New York. United States
| | - Li-Zhu Lin
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405. China
| |
Collapse
|
112
|
Xie T, Zhang Z, Zhang X, Qi C, Shen L, Peng Z. Appropriate PD-L1 Cutoff Value for Gastric Cancer Immunotherapy: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:646355. [PMID: 34540656 PMCID: PMC8440909 DOI: 10.3389/fonc.2021.646355] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 08/10/2021] [Indexed: 12/30/2022] Open
Abstract
Background Immunotherapy dramatically changed the treatment landscape of gastric cancer in recent years. PD-L1 expression was proposed as a biomarker; however, the treatment strategy according to PD-L1 is still uncertain. Here, we aimed to find the appropriate cutoff value of PD-L1 expression for gastric cancer immunotherapy. Methods We did a systematic electronic research of prospective clinical trials of gastric cancer immunotherapy across databases. Studies that provided subgroup analysis results stratified by PD-L1 expression were included. Objective response rate (ORR), disease control rate (DCR), hazard ratio (HR), and 95% confidential interval (CI) of progression-free survival (PFS) and overall survival (OS) at different PD-L1 cutoff values were extracted. Results Twelve studies and 6,488 patients in total were finally included for pooled analysis. ORR in allover, PD-L1-negative, combined positive score (CPS) ≥1, CPS ≥5, and CPS ≥10 population was 10%, 3%, 13%, 20%, and 23%, respectively. Immune checkpoint inhibitor (ICI) monotherapy failed to show survival advantage in allover and PD-L1-negative patients. Single-agent ICI therapy prolonged OS (HR = 0.84, 95% CI: 0.74–0.96) but not PFS (HR = 1.38, 95% CI: 0.91–2.09) in PD-L1 CPS ≥1 patients. For combined immunotherapy, ORR in allover, PD-L1-negative, CPS ≥1, CPS ≥5, and CPS ≥10 population was 64%, 57%, 48%, 60%, and 58%, respectively. Allover population could gain survival benefit from combined immunotherapy based on the results from Checkmate-649. OS (HR = 0.81, 95% CI: 0.71–0.92) and PFS (HR = 0.77, 95% CI: 0.69–0.86) were significantly prolonged in PD-L1 CPS ≥1 patients receiving combined immunotherapy. Conclusion Efficacy and survival advantages improved with PD-L1 CPS. CPS ≥1 was the cutoff value for ICI monotherapy to gain survival benefit. Combined immunotherapy prolonged PFS and OS in allover population but needs further study to confirm it.
Collapse
Affiliation(s)
- Tong Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhening Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
113
|
Topp BG, Thiagarajan K, De Alwis DP, Snyder A, Hellmann MD. Lesion-level heterogeneity of radiologic progression in patients treated with pembrolizumab. Ann Oncol 2021; 32:1618-1625. [PMID: 34543717 DOI: 10.1016/j.annonc.2021.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/20/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Disease progression is often considered a binary state reflecting presence or absence of response. Meaningful heterogeneity between metastatic sites of a given patient may exist, however, and may impact therapeutic outcomes. To characterize the heterogeneity of progression with immunotherapy, we evaluated lesion-level dynamics of pembrolizumab-treated patients across three tumor types. PATIENTS AND METHODS Individual metastatic lesion dynamics were analyzed retrospectively in patients with advanced melanoma, non-small-cell lung cancer (NSCLC), and gastric or gastroesophageal junction (G/GEJ) cancer who received pembrolizumab in KEYNOTE-001 or KEYNOTE-059. Primary progression was defined as radiologic progression as per RECIST v1.1 occurring at the first on-treatment study scan (∼9-12 weeks, +2-week window) and secondary progression as progression occurring beyond the first scan (∼14 weeks and beyond). The change in sum of target lesions and of individual lesions was examined, as were patterns and timing of progression. RESULTS 9239 individual lesions from 1194 patients were analyzed. Among patients with primary progression [39% (200/511) of patients with melanoma, 41% (179/432) with NSCLC, 61% (154/251) with G/GEJ cancer], most patients (51%-63%) had a mixture of growing, stable, and shrinking lesions. Despite overall primary progression, a minority of patients (19%-25%) had tumor growth at every metastatic site and 17%-32% had ≥1 shrinking lesion. Among patients with secondary progression [22% (113/511) of patients with melanoma, 27% (117/432) with NSCLC, 18% (44/251) with G/GEJ cancer], few patients had rebound growth (>20% increase in diameter from nadir) in all lesions whereas the majority (74%-84%) had sustained regression in ≥1 lesion. CONCLUSIONS Lesion-level heterogeneity at the time of disease progression was common in pembrolizumab-treated patients, with many patients demonstrating ongoing disease control in a subset of tumor sites. These results may inform clinical decision-making, trial design, and tumor sampling in the future.
Collapse
Affiliation(s)
- B G Topp
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc., Kenilworth, USA.
| | - K Thiagarajan
- Quantitative Systems Pharmacology, Vantage Research, Chennai, India
| | - D P De Alwis
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc., Kenilworth, USA
| | - A Snyder
- Global Clinical Development, Merck & Co., Inc., Kenilworth, USA
| | - M D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
114
|
Sun JM, Shen L, Shah MA, Enzinger P, Adenis A, Doi T, Kojima T, Metges JP, Li Z, Kim SB, Cho BC, Mansoor W, Li SH, Sunpaweravong P, Maqueda MA, Goekkurt E, Hara H, Antunes L, Fountzilas C, Tsuji A, Oliden VC, Liu Q, Shah S, Bhagia P, Kato K. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021; 398:759-771. [PMID: 34454674 DOI: 10.1016/s0140-6736(21)01234-4] [Citation(s) in RCA: 839] [Impact Index Per Article: 209.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND First-line therapy for advanced oesophageal cancer is currently limited to fluoropyrimidine plus platinum-based chemotherapy. We aimed to evaluate the antitumour activity of pembrolizumab plus chemotherapy versus chemotherapy alone as first-line treatment in advanced oesophageal cancer and Siewert type 1 gastro-oesophageal junction cancer. METHODS We did a randomised, placebo-controlled, double-blind, phase 3 study across 168 medical centres in 26 countries. Patients aged 18 years or older with previously untreated, histologically or cytologically confirmed, locally advanced, unresectable or metastatic oesophageal cancer or Siewert type 1 gastro-oesophageal junction cancer (regardless of PD-L1 status), measurable disease per Response Evaluation Criteria in Solid Tumors version 1.1, and Eastern Cooperative Oncology Group performance status of 0-1, were randomly assigned (1:1) to intravenous pembrolizumab 200 mg or placebo, plus 5-fluorouracil and cisplatin (chemotherapy), once every 3 weeks for up to 35 cycles. Randomisation was stratified by geographical region, histology, and performance status. Patients, investigators, and site staff were masked to group assignment and PD-L1 biomarker status. Primary endpoints were overall survival in patients with oesophageal squamous cell carcinoma and PD-L1 combined positive score (CPS) of 10 or more, and overall survival and progression-free survival in patients with oesophageal squamous cell carcinoma, PD-L1 CPS of 10 or more, and in all randomised patients. This trial is registered with ClinicalTrials.gov, NCT03189719, and is closed to recruitment. FINDINGS Between July 25, 2017, and June 3, 2019, 1020 patients were screened and 749 were enrolled and randomly assigned to pembrolizumab plus chemotherapy (n=373 [50%]) or placebo plus chemotherapy (n=376 [50%]). At the first interim analysis (median follow-up of 22·6 months), pembrolizumab plus chemotherapy was superior to placebo plus chemotherapy for overall survival in patients with oesophageal squamous cell carcinoma and PD-L1 CPS of 10 or more (median 13·9 months vs 8·8 months; hazard ratio 0·57 [95% CI 0·43-0·75]; p<0·0001), oesophageal squamous cell carcinoma (12·6 months vs 9·8 months; 0·72 [0·60-0·88]; p=0·0006), PD-L1 CPS of 10 or more (13·5 months vs 9·4 months; 0·62 [0·49-0·78]; p<0·0001), and in all randomised patients (12·4 months vs 9·8 months; 0·73 [0·62-0·86]; p<0·0001). Pembrolizumab plus chemotherapy was superior to placebo plus chemotherapy for progression-free survival in patients with oesophageal squamous cell carcinoma (6·3 months vs 5·8 months; 0·65 [0·54-0·78]; p<0·0001), PD-L1 CPS of 10 or more (7·5 months vs 5·5 months; 0·51 [0·41-0·65]; p<0·0001), and in all randomised patients (6·3 months vs 5·8 months; 0·65 [0·55-0·76]; p<0·0001). Treatment-related adverse events of grade 3 or higher occurred in 266 (72%) patients in the pembrolizumab plus chemotherapy group versus 250 (68%) in the placebo plus chemotherapy group. INTERPRETATION Compared with placebo plus chemotherapy, pembrolizumab plus chemotherapy improved overall survival in patients with previously untreated, advanced oesophageal squamous cell carcinoma and PD-L1 CPS of 10 or more, and overall survival and progression-free survival in patients with oesophageal squamous cell carcinoma, PD-L1 CPS of 10 or more, and in all randomised patients regardless of histology, and had a manageable safety profile in the total as-treated population. FUNDING Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Jong-Mu Sun
- Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea.
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Manish A Shah
- Weill Cornell Medical College, New York City, NY, USA
| | | | - Antoine Adenis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm, Université Montpellier, ICM, Montpellier, France
| | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Jean-Philippe Metges
- CHU Brest-Institut de Cancerologie et d'Hematologie ARPEGO Network, Brest, France
| | - Zhigang Li
- Section of Oesophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sung-Bae Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Shau-Hsuan Li
- Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | - Eray Goekkurt
- Hematology Oncology Practice Eppendorf and University Cancer Center Hamburg, Hamburg, Germany
| | | | - Luis Antunes
- University Hospital of Santa Maria, Federal University of Santa Maria, and Viver Research Center, Santa Maria, Brazil
| | | | | | | | - Qi Liu
- Merck, Kenilworth, NJ, USA
| | | | | | - Ken Kato
- National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
115
|
Van Cutsem E, Valderrama A, Bang YJ, Fuchs CS, Shitara K, Janjigian YY, Qin S, Larson TG, Shankaran V, Stein S, Norquist JM, Kher U, Shah S, Alsina M. Quality of life with first-line pembrolizumab for PD-L1-positive advanced gastric/gastroesophageal junction adenocarcinoma: results from the randomised phase III KEYNOTE-062 study. ESMO Open 2021; 6:100189. [PMID: 34371381 PMCID: PMC8358416 DOI: 10.1016/j.esmoop.2021.100189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND In the randomised phase III KEYNOTE-062 study, pembrolizumab was non-inferior to chemotherapy for overall survival in patients with programmed death-ligand 1 (PD-L1)-positive [combined positive score (CPS) ≥1] advanced gastric/gastroesophageal junction (GEJ) cancer. We present findings of prespecified health-related quality-of-life (HRQOL) analyses for pembrolizumab versus chemotherapy in this population. MATERIALS AND METHODS HRQOL, a secondary endpoint, was measured in patients who received ≥1 dose of study treatment and completed ≥1 HRQOL questionnaire [European Organisation for the Research and Treatment of Cancer (EORTC) 30-question quality-of-life (QLQ-C30), EORTC 22-question quality-of-life gastric-cancer-specific module (QLQ-STO22)]. Least squares mean (LSM) change (baseline to week 18) in global health status/quality of life (GHS/QOL; EORTC QLQ-C30) and time to deterioration (TTD) in GHS/QOL, nausea/vomiting and appetite loss scores (EORTC QLQ-C30) and abdominal pain/discomfort scores (EORTC QLQ-STO22) were evaluated. RESULTS The HRQOL population comprised 495 patients with CPS ≥1 (pembrolizumab, 252; chemotherapy, 243). Compliance rates at week 18 were similar for pembrolizumab and chemotherapy (EORTC QLQ-C30, 87.9% and 81.9%; EORTC QLQ-STO22, 87.9% and 81.3%, respectively). There was no between-arm difference in LSM score change in GHS/QOL [-0.16; 95% confidence interval (CI) -5.01 to 4.69; P = 0.948]. The LSM score change for most subscales showed comparable worsening in both arms. TTD for GHS/QOL [hazard ratio (HR), 0.96; 95% CI, 0.67-1.38; P = 0.826], appetite loss (HR, 0.83; 95% CI, 0.58-1.20; P = 0.314) and pain (HR, 1.22; 95% CI, 0.78-1.91; P = 0.381) were similar between arms. Longer TTD was observed for pembrolizumab versus chemotherapy for nausea/vomiting (HR, 0.61; 95% CI, 0.44-0.85; P = 0.003). CONCLUSIONS HRQOL was maintained with first-line treatment with pembrolizumab in patients with PD-L1-positive advanced gastric/GEJ cancer and was similar between pembrolizumab and chemotherapy in this population.
Collapse
Affiliation(s)
- E Van Cutsem
- Department of Digestive Oncology, University Hospital Gasthuisberg Leuven and KU Leuven, Leuven, Belgium.
| | - A Valderrama
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, USA
| | - Y-J Bang
- Department of Biomedical Research, Seoul National University College of Medicine, Seoul, South Korea
| | - C S Fuchs
- Department of Internal Medicine: Hematology, Medical Oncology, Gastro-oncology, Yale University Cancer Center, Smilow Cancer Hospital, New Haven, USA
| | - K Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Y Y Janjigian
- Department of Gastrointestinal Oncology, Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - S Qin
- Cancer Center, PLA Cancer Centre of Nanjing Bayi Hospital, Nanjing, China
| | - T G Larson
- Department of Hematology/Oncology, Minnesota Oncology Hematology, Minneapolis
| | - V Shankaran
- Department of Medical Oncology, Seattle Cancer Care Alliance, Seattle
| | - S Stein
- Department of Internal Medicine: Hematology, Medical Oncology, Gastro-oncology, Yale University Cancer Center, Smilow Cancer Hospital, New Haven, USA
| | - J M Norquist
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, USA
| | - U Kher
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, USA
| | - S Shah
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, USA
| | - M Alsina
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology, Barcelona; University Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
116
|
Hu J, Yang S, Wang J, Zhang Q, Zhao L, Zhang D, Yu D, Jin M, Ma H, Liu H, Xue J, Zhang T. Blood alkaline phosphatase predicts prognosis of patients with advanced HER2-negative gastric cancer receiving immunotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1316. [PMID: 34532453 PMCID: PMC8422101 DOI: 10.21037/atm-21-3376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/11/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND Immune checkpoint blockade is effective against many cancer types, but few patients achieve a complete response (OR). Therefore, effective prognostic biomarkers are needed for metastatic gastric cancer (GC) patients after immune treatment. The present study assessed the value of hematological parameters as markers of the effectiveness of immune checkpoint blockade among metastatic GC patients. METHODS This retrospective study included patients with metastatic GC who underwent multiline chemotherapy including at least two courses of immunotherapy between September 2018 and December 2020. Patient and tumor characteristics were tested for prognostic significance by analysis of variance or chi-square test. Kaplan-Meier and Cox analyses were performed to identify factors associated with progression-free survival (PFS). RESULTS Sixty-one GC patients (mean age 55.61±11.97 years, range 23-80 years, 24 females, and 37 males) were included, and 27, 9 and 25 cases had organ only, peritoneum only, and simultaneous organ and peritoneum metastasis, respectively. Gastrectomy was performed in 24 cases, and there was no operative treatment in the other 37 cases, while all patients received two or more lines of chemotherapy. After immune treatment, 13 patients achieved a partial response (PR), 16 stable disease (SD), and 32 progressive disease (PD). The median PFS was 4.93±3.47 months. An alkaline phosphatase (ALP) level >225 U/L, a lactate dehydrogenase level (LDH) >299 U/L, and a body mass index (BMI) >24 kg/m2 were associated with a short PFS (P=0.01, P=0.008, and P=0.039, respectively). A Cox multivariate proportional hazard model indicated that higher ALP level was a significant prognostic indicator for adverse PFS. CONCLUSIONS Our data show an ALP cutoff of 225 U/L offered good prognostic sensitivity for HER2-negative metastatic GC. ALP measurement represents a convenient, cost-effective, and relatively sensitive screening tool, and prospective studies involving its evaluation in addition to other biomarkers in metastatic GC patients are indicated.
Collapse
Affiliation(s)
- Jianli Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengli Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyue Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dejun Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xue
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
117
|
Tabernero J, Bang YJ, Van Cutsem E, Fuchs CS, Janjigian YY, Bhagia P, Li K, Adelberg D, Qin SK. KEYNOTE-859: a Phase III study of pembrolizumab plus chemotherapy in gastric/gastroesophageal junction adenocarcinoma. Future Oncol 2021; 17:2847-2855. [PMID: 33975465 PMCID: PMC9892960 DOI: 10.2217/fon-2021-0176] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/30/2021] [Indexed: 01/19/2023] Open
Abstract
Current guidelines recommend two-drug cytotoxic chemotherapy with a fluoropyrimidine (fluorouracil or capecitabine) and a platinum-based agent (oxaliplatin or cisplatin) as first-line treatment for advanced gastric cancer. Pembrolizumab monotherapy has demonstrated durable antitumor activity in patients with advanced programmed death ligand 1-positive (combined positive score ≥1) gastric/gastroesophageal junction adenocarcinoma. Accumulating evidence indicates that combining pembrolizumab with standard-of-care chemotherapy for the treatment of advanced or metastatic cancer improves clinical outcomes. We describe the rationale for and the design of the randomized, double-blind, placebo-controlled, Phase III KEYNOTE-859 study, which is investigating pembrolizumab in combination with chemotherapy as first-line treatment for patients with human epidermal growth factor receptor 2-negative advanced unresectable or metastatic gastric/gastroesophageal junction adenocarcinoma. The planned sample size is 1542 patients, and the primary end point is overall survival. Clinical trial registration: NCT03675737 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Josep Tabernero
- Vall d'Hebron Hospital Campus & Institute of Oncology, UVic-UCC, IOB-Quiron, 08035, Barcelona, Spain
| | - Yung-Jue Bang
- Seoul National University College of Medicine, 103 Daehak-ro, Ihwa-dong, Jongno-gu, Seoul, South Korea
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven & KU Leuven, Leuven, Belgium
| | - Charles S Fuchs
- Yale Cancer Center & Smilow Cancer Hospital, New Haven, CT 06511, USA
| | | | | | - Kan Li
- Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | | | - Shu Kui Qin
- PLA Cancer Centre of Nanjing, Jinling Hospital, Nanjing, 34210002, China
| |
Collapse
|
118
|
Liang C, Wu HM, Yu WM, Chen W. Research status on immunotherapy trials of gastric cancer. World J Clin Cases 2021; 9:5782-5793. [PMID: 34368297 PMCID: PMC8316931 DOI: 10.12998/wjcc.v9.i21.5782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/13/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
The breakthrough of immune checkpoint inhibitor (ICI) therapy has created extensive opportunities for cancer immunotherapy. Especially, the block of programmed death-1/programmed death ligand 1 (PD-L1) axis using ICIs has become a new therapeutic strategy to treat advanced gastric cancer (GC). However, in the past decade, single-arm and randomized trials for single-drug ICI therapy showed that the therapeutic effect was not satisfactory, including clinical trials for advanced GC. However, after selecting suitable predictive biomarkers and developing a combination of anti-angiogenic targeted drugs and other chemotherapeutic drugs, the objective response rate and progression-free survival of patients with gastric cancer were improved significantly. The United States Food and Drug Administration has approved treatment with pembrolizumab for patients with advanced GC with PD-L1 expression or microsatellite instability-high/mismatch repair deficiency. In this review, the updated data from the latest trial results of combination immunotherapy for GC are presented. Based on the outcome of combination therapy, we discuss its possible molecular mechanism and summarize effective predictive biomarkers. We also discuss possible problems stemming from results of other clinical trials of ICI treatment and propose other directions for ICI therapy.
Collapse
Affiliation(s)
- Chao Liang
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo 315040, Zhejiang Province, China
| | - Heng-Miao Wu
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Wei-Ming Yu
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo 315040, Zhejiang Province, China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| |
Collapse
|
119
|
Regional Delivery of Anti-PD-1 Agent for Colorectal Liver Metastases Improves Therapeutic Index and Anti-Tumor Activity. Vaccines (Basel) 2021; 9:vaccines9080807. [PMID: 34451932 PMCID: PMC8402391 DOI: 10.3390/vaccines9080807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/26/2022] Open
Abstract
Metastatic liver tumors have presented challenges with the use of checkpoint inhibitors (CPIs), with only limited success. We hypothesize that regional delivery (RD) of CPIs can improve activity in the liver and minimize systemic exposure, thereby reducing immune-related adverse events (irAE). Using a murine model of colorectal cancer liver metastases (LM), we confirmed high levels of PD-L1 expression on the tumor cells and liver myeloid-derived suppressor cells (L-MDSC). In vivo, we detected improved LM response at 3 mg/kg on PTD7 via portal vein (PV) regional delivery as compared to 3 mg/kg via tail vein (TV) systemic delivery (p = 0.04). The minimal effective dose at PTD7 was 5 mg/kg (p = 0.01) via TV and 0.3 mg/kg (p = 0.02) via PV. We detected 6.7-fold lower circulating CPI antibody levels in the serum using the 0.3 mg/kg PV treatment compared to the 5 mg/kg TV cohort (p < 0.001) without increased liver toxicity. Additionally, 3 mg/kg PV treatment resulted in increased tumor cell apoptotic signaling compared to 5 mg/kg TV (p < 0.05). Therefore, RD of an anti-PD-1 CPI therapy for CRCLM may improve the therapeutic index by reducing the total dose required and limiting the systemic exposure. These advantages could expand CPI indications for liver tumors.
Collapse
|
120
|
Kim N, Yu JI, Lim DH, Lee J, Kim ST, Hong JY, Kang WK, Jeong WK, Kim KM. Prognostic Impact of Sarcopenia and Radiotherapy in Patients With Advanced Gastric Cancer Treated With Anti-PD-1 Antibody. Front Immunol 2021; 12:701668. [PMID: 34305941 PMCID: PMC8298191 DOI: 10.3389/fimmu.2021.701668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND We explored the combined effects of sarcopenia (SAR) and radiotherapy (RT) on outcomes in patients with advanced gastric cancer (AGC) treated with immune-checkpoint blockade (ICB). METHODS Among 185 patients with AGC treated with ICB, we defined SAR as skeletal muscle index <49 cm2/m2 for men and <31 cm2/m2 for women; 93 patients met criteria. We defined high neutrophil-to-lymphocyte ratio (hNLR) as NLR≥3. Palliative RT was performed in 37 patients (20%) before ICB. RESULTS We frequently observed hNLR in patients with SAR (53% vs. 35%, p = 0.02). The median overall survival (OS) for the entire cohort was 5 months. Stratification by risk factors of SAR or hNLR revealed a significant difference in median OS (0 [N = 60] vs. 1 [N = 76] vs. 2 [N = 49]: 7.6 vs. 6.4 vs. 2.2 months, p < 0.001). Patients with microsatellite instability-high (MSI-H, N = 19) or Epstein-Barr virus (EBV)-positive tumors (N = 13) showed favorable outcomes compared to those with microsatellite stable (MSS, N = 142) tumors (median OS, not reached vs. 16.8 vs. 3.8 months, respectively). The benefit of RT was evident in patients with both SAR and hNLR (median OS, 3.1 vs. 1.3 months, p = 0.02) and MSS/EBV-negative tumor (median OS, 6.5 vs. 3.5 months, p = 0.03), but outcomes after RT in MSI-H tumor were not significantly different. In multivariable analysis, SAR/hNLR, molecular subtypes, and a history of RT were associated with OS (all p < 0.05). CONCLUSIONS We demonstrated the negative predictive value of SAR/hNLR on outcomes after ICB for AGC, and the history of RT could overcome the negative impact of SAR/hNLR and the MSS/EBV-negative subtype.
Collapse
Affiliation(s)
- Nalee Kim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong Il Yu
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeeyun Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seung Tae Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung Yong Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Won Ki Kang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Woo Kyoung Jeong
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyoung-Mee Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
121
|
Rzhevskiy A, Kapitannikova A, Malinina P, Volovetsky A, Aboulkheyr Es H, Kulasinghe A, Thiery JP, Maslennikova A, Zvyagin AV, Ebrahimi Warkiani M. Emerging role of circulating tumor cells in immunotherapy. Theranostics 2021; 11:8057-8075. [PMID: 34335980 PMCID: PMC8315079 DOI: 10.7150/thno.59677] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022] Open
Abstract
Over the last few years, immunotherapy, in particular, immune checkpoint inhibitor therapy, has revolutionized the treatment of several types of cancer. At the same time, the uptake in clinical oncology has been slow owing to the high cost of treatment, associated toxicity profiles and variability of the response to treatment between patients. In response, personalized approaches based on predictive biomarkers have emerged as new tools for patient stratification to achieve effective immunotherapy. Recently, the enumeration and molecular analysis of circulating tumor cells (CTCs) have been highlighted as prognostic biomarkers for the management of cancer patients during chemotherapy and for targeted therapy in a personalized manner. The expression of immune checkpoints on CTCs has been reported in a number of solid tumor types and has provided new insight into cancer immunotherapy management. In this review, we discuss recent advances in the identification of immune checkpoints using CTCs and shed light on the potential applications of CTCs towards the identification of predictive biomarkers for immunotherapy.
Collapse
Affiliation(s)
- Alexey Rzhevskiy
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Institute for Urology and Reproductive Health, Sechenov University, Moscow 119991, Russia
| | - Alina Kapitannikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Polina Malinina
- Privolzhsky Research Medical University, 10/1, Minini Pozharsky Square, Nizhny Novgorod 603005, Russia
| | - Arthur Volovetsky
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russia
| | | | - Arutha Kulasinghe
- Queensland University of Technology, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Woolloongabba, QLD 4102, Australia
- Translational Research Institute, Woolloongabba, QLD 4102 Australia
| | - Jean Paul Thiery
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Anna Maslennikova
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russia
- The Chair of Cancer, Radiotherapy and Radiologic Diagnostics, Privolzhsky Research Medical University, Nizhniy Novgorod. Russia 603005
| | - Andrei V. Zvyagin
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- IBCh - Shemyakin Ovchinnikov Institute of BioOrganic Chemistry of the Russian Academy of Sciences, Miklukho Maklai Street, 16, Moscow, Russia
| | - Majid Ebrahimi Warkiani
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- School of Biomedical Engineering, University of Technology Sydney, 2007 Sydney, Australia
| |
Collapse
|
122
|
Cho HJ, Kim WR, Kim JH, Kim DH, Kim DJ, Kang H. Colonic Perforation After Treatment With Nivolumab in Esophageal Cancer: A Case Report. Ann Coloproctol 2021; 37:S39-S43. [PMID: 34167190 PMCID: PMC8359691 DOI: 10.3393/ac.2020.00213.0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023] Open
Abstract
With the advent of checkpoint inhibitors, it has opened up opportunities for numerous cancer patients. However, as is the case with every treatment, complications need to be weighed. Gastrointestinal adverse effects, such as diarrhea and colitis are well-known complications for checkpoint inhibitors. In severe cases, colitis-induced colonic perforation may occur with an estimation of 1.0% to 1.5% in anti-CTLA-4 antibodies. However, only a handful of cases of such devastating complications have been reported in anti-PD-1 antibodies such as pembrolizumab and nivolumab. We here report a case of intestinal perforation in a patient treated with nivolumab.
Collapse
Affiliation(s)
- Hye Jung Cho
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Woo Ram Kim
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Joo-Hang Kim
- Department of Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Duk Hwan Kim
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Dae Jung Kim
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Haeyoun Kang
- Department of Pathology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
123
|
Abstract
Gastric cancer represents a major global health problem. Approximately half of patients are diagnosed with early stage disease and surgical resection is potentially curative. The addition of combination chemotherapy, with or without radiotherapy, to surgery has been shown to improve outcomes. In metastatic disease, combination chemotherapy in the form of 2- or 3-drug regimens has been used. The aim of this chapter is to summarize currently approved systemic treatment options for gastric cancer and to highlight several promising treatments currently under investigation.
Collapse
Affiliation(s)
- Andrew Hsu
- The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital/The Miriam Hospital, 164 Summit Avenue, Fain Building, Third Floor, Providence, RI 02906, USA
| | - Alexander G Raufi
- The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital/The Miriam Hospital, 164 Summit Avenue, Fain Building, Third Floor, Providence, RI 02906, USA.
| |
Collapse
|
124
|
Peng Z, Wei J, Wang F, Ying J, Deng Y, Gu K, Cheng Y, Yuan X, Xiao J, Tai Y, Wang L, Zou J, Zhang Y, Shen L. Camrelizumab Combined with Chemotherapy Followed by Camrelizumab plus Apatinib as First-line Therapy for Advanced Gastric or Gastroesophageal Junction Adenocarcinoma. Clin Cancer Res 2021; 27:3069-3078. [PMID: 33766817 DOI: 10.1158/1078-0432.ccr-20-4691] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/28/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Capecitabine plus oxaliplatin (CAPOX) is one of the standard first-line treatments for unresectable, advanced, or metastatic gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. Camrelizumab shows promising antitumor activity in advanced or metastatic G/GEJ adenocarcinoma in a phase I study. We reported the outcomes of cohort 1 in a multicenter, open-label, phase II trial, which assessed camrelizumab in combination with CAPOX followed by camrelizumab plus apatinib as a first-line combination regimen for advanced or metastatic G/GEJ adenocarcinoma. PATIENTS AND METHODS Systemic treatment-naïve patients with EGFR2-negative advanced or metastatic G/GEJ adenocarcinoma received initial camrelizumab plus CAPOX for 4-6 cycles, and patients without progressive disease were administrated subsequent camrelizumab plus apatinib. Primary endpoint was objective response rate (ORR). RESULTS All 48 enrolled patients comprised the efficacy and safety analysis population. The ORR was 58.3% [95% confidence interval (CI), 43.2-72.4] with this combination regimen. Median duration of response was 5.7 months (95% CI, 4.4-8.3). Median overall survival was 14.9 months (95% CI, 13.0-18.6), and median progression-free survival was 6.8 months (95% CI, 5.6-9.5), respectively. The most common grade ≥3 treatment-related adverse events (>10%) were decreased platelet count (20.8%), decreased neutrophil count (18.8%), and hypertension (14.6%). Treatment-related death occurred in 1 patient (2.1%) due to abnormal hepatic function and interstitial lung disease. CONCLUSIONS Camrelizumab combined with CAPOX followed by camrelizumab plus apatinib demonstrated encouraging antitumor activity and manageable toxicity as first-line therapy for patients with advanced or metastatic G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Zhi Peng
- Department of GI Oncology, Beijing Cancer Hospital, Beijing, P.R. China
| | - Jia Wei
- Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Jieer Ying
- Department of Abdominal Oncology, Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | - Yanhong Deng
- Department of Medical Oncology (Department of Chemotherapy), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Kangsheng Gu
- Department of Medical Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
| | - Ying Cheng
- Department of Medical Oncology, Jilin Cancer Hospital, Changchun, P.R. China
| | - Xianglin Yuan
- Oncology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Juxiang Xiao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yanfei Tai
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, P.R. China
| | - Linna Wang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, P.R. China
| | - Jianjun Zou
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, P.R. China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, P.R. China.
| | - Lin Shen
- Department of GI Oncology, Beijing Cancer Hospital, Beijing, P.R. China.
| |
Collapse
|
125
|
Barchi LC, Ramos MFKP, Dias AR, Forones NM, Carvalho MPD, Castro OAP, Kassab P, Costa-Júnior WLD, Weston AC, Zilberstein B, Ferraz ÁAB, ZeideCharruf A, Brandalise A, Silva AMD, Alves B, Marins CAM, Malheiros CA, Leite CV, Bresciani CJC, Szor D, Mucerino DR, Wohnrath DR, JirjossIlias E, Martins Filho ED, PinatelLopasso F, Coimbra FJF, Felippe FEC, Tomasisch FDS, Takeda FR, Ishak G, Laporte GA, Silva HJT, Cecconello I, Rodrigues JJG, Grande JCD, Lourenço LG, Motta LMD, Ferraz LR, Moreira LF, Lopes LR, Toneto MG, Mester M, Rodrigues MAG, Franciss MY, AdamiAndreollo N, Corletta OC, Yagi OK, Malafaia O, Assumpção PP, Savassi-Rocha PR, Colleoni Neto R, Oliveira RJD, AissarSallun RA, Weschenfelder R, Oliveira SCVD, Abreu TBD, Castria TBD, Ribeiro Junior U, Barra W, Freitas Júnior WRD. BRAZILIAN GASTRIC CANCER ASSOCIATION GUIDELINES (PART 2): UPDATE ON TREATMENT. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2021; 34:e1563. [PMID: 34008707 PMCID: PMC8121052 DOI: 10.1590/0102-672020210001e1563] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND : The II Brazilian Consensus on Gastric Cancer of the Brazilian Gastric Cancer Association BGCA (Part 1) was recently published. On this occasion, countless specialists working in the treatment of this disease expressed their opinion in the face of the statements presented. AIM : To present the BGCA Guidelines (Part 2) regarding indications for surgical treatment, operative techniques, extension of resection and multimodal treatment. METHODS To formulate these guidelines, the authors carried out an extensive and current review regarding each declaration present in the II Consensus, using the Medline/PubMed, Cochrane Library and SciELO databases initially with the following descriptors: gastric cancer, gastrectomy, lymphadenectomy, multimodal treatment. In addition, each statement was classified according to the level of evidence and degree of recommendation. RESULTS : Of the 43 statements present in this study, 11 (25,6%) were classified with level of evidence A, 20 (46,5%) B and 12 (27,9%) C. Regarding the degree of recommendation, 18 (41,9%) statements obtained grade of recommendation 1, 14 (32,6%) 2a, 10 (23,3%) 2b e one (2,3%) 3. CONCLUSION : The guidelines complement of the guidelines presented here allows surgeons and oncologists who work to combat gastric cancer to offer the best possible treatment, according to the local conditions available.
Collapse
Affiliation(s)
- Leandro Cardoso Barchi
- Hospital das Clinicas HCFMUSP, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
- Faculty of Medicine São Leopoldo Mandic, Campinas, SP, Brazil
| | | | - André Roncon Dias
- Hospital das Clinicas HCFMUSP, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | - Paulo Kassab
- Department of Surgery, Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Wilson Luiz da Costa-Júnior
- Department of Abdominal Surgery, AC Camargo Cancer Center, São Paulo, SP, Brazil
- Department of Medicine, Baylor College of Medicine, Houston,Texas
| | | | - Bruno Zilberstein
- Hospital das Clinicas HCFMUSP, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
- Faculty of Medicine São Leopoldo Mandic, Campinas, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Zhang ML, Deshpande V. Histopathology of Gastrointestinal Immune-related Adverse Events: A Practical Review for the Practicing Pathologist. Am J Surg Pathol 2021; 46:e15-e26. [PMID: 33999556 DOI: 10.1097/pas.0000000000001730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Immune checkpoint inhibitors target checkpoint proteins with the goal of reinvigorating the host immune system and thus restoring antitumor response. With the dramatic increase in the use of checkpoint inhibitors for cancer treatment, surgical pathologists have assumed a major role in predicting the therapeutic efficacy (score based on programmed cell death ligand 1 immunohistochemistry and mismatch repair protein loss) as well as diagnosing the complications associated with these medications. Immune-related adverse events (irAEs) manifest as histologic changes seen in both the upper and lower gastrointestinal tract, and when viewed in isolation, may be morphologically indistinguishable from a wide range of diseases including infections, celiac disease, and inflammatory bowel disease, among others. Evaluation of biopsies from both the upper and lower gastrointestinal tract can aid in the distinction of gastrointestinal irAEs from their mimics. In the liver, the histologic changes of hepatic irAEs overlap with de novo diseases associated with hepatitic and cholangitic patterns of injury. The diagnosis of irAEs requires communication and collaboration from the pathologist, oncologist, and gastroenterologist. This review provides a background framework and illustrates the histologic features and differential diagnosis of gastrointestinal and hepatic irAEs.
Collapse
Affiliation(s)
- M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | | |
Collapse
|
127
|
Ouyang T, Cao Y, Kan X, Chen L, Ren Y, Sun T, Yan L, Xiong B, Liang B, Zheng C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front Oncol 2021; 11:621639. [PMID: 34046338 PMCID: PMC8144509 DOI: 10.3389/fonc.2021.621639] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immune Checkpoint Inhibitors (ICI) have been progressively used in cancer treatment and produced unique toxicity profiles. This systematic review aims to comprehend the patterns and occurrence of treatment-related adverse events (trAEs) based on ICI. Methods PICOS/PRISMA methods were used to identify published English-language on PubMed, Web of Science, and Scopus from 2015 to 2020. Published clinical trials on ICI monotherapy, combined ICIs, and ICI plus other treatment with tabulated data on grade≥3 trAEs were included. Odds ratio (OR), χ2 tests were used to analyze for effect size and associations. Results This review included 145 clinical trials involving 21786 patients. Grade 3-5 trAEs were more common with ICI when they were plused with other treatments compared with ICI monotherapy(54.3% versus 17.7%, 46.1%, p<0.05). Grade 3-5 trAEs were also more common with CTLA-4 mAbs compared with anti-PD-1 and anti-PD-L1 (34.2% versus 15.1%, 13.6%, p<0.05). Hyperthyroidism (OR 3.8, 95%CI 1.7–8.6), nausea (OR 3.7, 95%CI 2.5–5.3), diarrhea (OR 2.7, 95%CI 2.2–3.2), colitis (OR 3.4, 95%CI 2.7–4.3), ALT increase (OR 4.9, 95%CI 3.9–6.1), AST increase (OR 3.8, 95%CI 3.0–4.9), pruritus (OR 2.4, 95%CI 1.5–3.9), rash (OR 2.8, 95%CI 2.1–3.8), fatigue (OR 2.8, 95%CI 2.2–3.7), decreased appetite (OR 2.4, 95%CI 1.5–3.8), and hypophysitis (OR 2.0, 95%CI 1.2–3.3) were more frequent with combined ICIs. Diarrhea (OR 8.1, 95%CI 6.4–10.3), colitis (OR 12.2, 95%CI 8.7–17.1), ALT increase (OR 5.1, 95%CI 3.5–7.4), AST increase (OR 4.2, 95%CI 2.8–6.3), pruritus (OR 4.1, 95%CI 2.0–8.4), rash (OR 4.4, 95%CI 2.9–6.8), hypophysitis (OR 12.1, 95%CI 6.3–23.4) were more common with CTLA-4 mAbs; whereas pneumonitis (OR 4.7, 95% CI 2.1–10.3) were more frequent with PD-1 mAbs. Conclusions Different immune checkpoint inhibitors are associated with different treatment-related adverse events profiles. A comprehensive data in this systematic review will provide comprehensive information for clinicians.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Liangliang Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
128
|
Kim JW. Gastrointestinal cancer treatment with immune checkpoint inhibitors. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2021. [DOI: 10.5124/jkma.2021.64.5.342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Immuno-oncological treatment approaches, particularly with the use of immune checkpoint inhibitors such as antiprogrammed death 1 (PD-1)/programmed death ligand 1 antibody or anti-cytotoxic T-lymphocyte associated protein 4 antibody, have become the standard treatment for gastrointestinal cancers. However, gastrointestinal cancers show an overall modest tumor response to immune checkpoint inhibitors. Nevertheless, subgroups such as tumors that are DNA mismatch repair-deficient or have high microsatellite instability particularly benefit from immune checkpoint inhibitors. Even in the first-line setting for colorectal cancer, the clinical efficacy of pembrolizumab, an anti–PD-1 antibody, was superior to that of chemotherapy. Recently, a combination of atezolizumab, an anti-programmed death ligand 1 antibody, and bevacizumab was approved as the first-line treatment for hepatocellular carcinoma, and was reported as superior to sorafenib. Nivolumab, an anti–PD-1 antibody that is added to chemotherapy as the first-line treatment for gastric cancer, resulted in longer survival compared with chemotherapy alone. Further studies are ongoing to investigate additional immune checkpoint inhibitors for other gastrointestinal cancers. This review aims to provide an overview of the results of clinical trials for immune checkpoint inhibitors in gastrointestinal cancers, including colorectal cancer, gastric cancer, hepatocellular carcinoma, pancreatic cancer, and biliary tract cancer.
Collapse
|
129
|
Dolan DP, Swanson SJ. The modern approach to esophagectomy-review of the shift towards minimally invasive surgery. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:901. [PMID: 34164535 PMCID: PMC8184437 DOI: 10.21037/atm.2020.03.143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The treatment of esophageal cancer has significantly advanced in the last 10 years and now includes multimodal treatment with a continued emphasis on surgical management. Minimally invasive esophagectomy (MIE) has been performed for almost 25 years and, in comparison to open esophagectomy techniques, MIE has shown to be equivalent or better in terms of its perioperative and oncologic outcomes. This paper reviews the evidence for MIE and recommends it should be offered as the first approach for esophagectomy surgery in the modern era.
Collapse
Affiliation(s)
- Daniel P Dolan
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott J Swanson
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
130
|
Fu J, Li WZ, McGrath NA, Lai CW, Brar G, Xiang YQ, Xie C. Immune Checkpoint Inhibitor Associated Hepatotoxicity in Primary Liver Cancer Versus Other Cancers: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:650292. [PMID: 33968750 PMCID: PMC8097087 DOI: 10.3389/fonc.2021.650292] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Overall risks of hepatotoxicity with immune checkpoint inhibitors (ICIs) have yet to be compared in primary liver cancers to other solid tumors. METHODS We reviewed data from the PubMed, Embase, and Scopus databases, and assessed the risk of hepatotoxicity associated with ICIs. RESULTS A total of 117 trials were eligible for the meta-analysis, including 7 trials with primary liver cancers. The most common hepatotoxicity was ALT elevation (incidence of all grade 5.29%, 95% CI 4.52-6.20) and AST elevation (incidence of all grade 5.88%, 95% CI 4.96-6.97). The incidence of all grade ALT and AST elevation was 6.01% and 6.84% for anti-PD-1 (95% CI 5.04-7.18/5.69-8.25) and 3.60% and 3.72% for anti-PD-L1 (95% CI 2.72-4.76/2.82-4.94; p< 0.001/p<0.001). The incidence of ≥ grade 3 ALT and AST elevation was 1.54% and 1.48% for anti-PD-1 (95% CI 1.19-1.58/1.07-2.04) and 1.03% and 1.08% for anti-PD-L1 (95% CI 0.71-1.51/0.80-1.45; p= 0.002/p<0.001). The incidence of all grade ALT and AST elevation was 13.3% and 14.2% in primary liver cancers (95% CI 11.1-16.0 and 9.93-20.36) vs. 4.92% and 5.38% in other solid tumors (95% CI 4.21-5.76 and 4.52-5.76 in other solid tumors; p <0.001/p<0.001). CONCLUSION Our study indicates that anti-PD-1 is associated with a higher risk of all- and high-grade hepatotoxicity compared to anti-PD-L1, and primary liver cancers are associated with a higher risk of all- and high-grade hepatotoxicity compared to other solid tumors.
Collapse
Affiliation(s)
- Jianyang Fu
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wang-Zhong Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Nicole A. McGrath
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Chunwei Walter Lai
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Gagandeep Brar
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Changqing Xie
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
131
|
Bone Metastases from Gastric Cancer: What We Know and How to Deal with Them. J Clin Med 2021; 10:jcm10081777. [PMID: 33921760 PMCID: PMC8073984 DOI: 10.3390/jcm10081777] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/28/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is the third cause of cancer-related death worldwide; the prognosis is poor especially in the case of metastatic disease. Liver, lymph nodes, peritoneum, and lung are the most frequent sites of metastases from GC; however, bone metastases from GC have been reported in the literature. Nevertheless, it is unclear how the metastatic sites may affect the prognosis. In particular, knowledge about the impact of bone metastases on GC patients’ outcome is scant, and this may be related to the rarity of bone lesions and/or their underestimation at the time of diagnosis. In fact, there is still a lack of specific recommendation for their detection at the diagnosis. Then, the majority of the evidences in this field came from retrospective analysis on very heterogeneous study populations. In this context, the aim of this narrative review is to delineate an overview about the evidences existing about bone metastases in GC patients, focusing on their incidence and biology, the prognostic role of bone involvement, and their possible implication in the treatment choice.
Collapse
|
132
|
Moehler M, Dvorkin M, Boku N, Özgüroğlu M, Ryu MH, Muntean AS, Lonardi S, Nechaeva M, Bragagnoli AC, Coşkun HS, Cubillo Gracian A, Takano T, Wong R, Safran H, Vaccaro GM, Wainberg ZA, Silver MR, Xiong H, Hong J, Taieb J, Bang YJ. Phase III Trial of Avelumab Maintenance After First-Line Induction Chemotherapy Versus Continuation of Chemotherapy in Patients With Gastric Cancers: Results From JAVELIN Gastric 100. J Clin Oncol 2021; 39:966-977. [PMID: 33197226 PMCID: PMC8078426 DOI: 10.1200/jco.20.00892] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The role of maintenance therapy for gastric (GC) or gastroesophageal junction cancer (GEJC) is unclear. We investigated avelumab (anti-programmed death ligand-1 [PD-L1]) maintenance after first-line induction chemotherapy for GC/GEJC. PATIENTS AND METHODS JAVELIN Gastric 100 was a global, open-label, phase III trial. Eligible patients had untreated, unresectable, human epidermal growth factor receptor 2-negative, locally advanced or metastatic GC or GEJC. Patients without progressive disease after 12 weeks of first-line chemotherapy with oxaliplatin plus a fluoropyrimidine were randomly assigned 1:1 to avelumab 10 mg/kg every 2 weeks or continued chemotherapy, stratified by region (Asia v non-Asia). The primary end point was overall survival (OS) after induction chemotherapy in all randomly assigned patients or the PD-L1-positive randomly assigned population (≥ 1% of tumor cells; 73-10 assay). RESULTS A total of 805 patients received induction; 499 were randomly assigned to avelumab (n = 249) or continued chemotherapy (n = 250). Median OS was 10.4 months (95% CI, 9.1 to 12.0 months) versus 10.9 months (95% CI, 9.6 to 12.4 months) and 24-month OS rate was 22.1% versus 15.5% with avelumab versus chemotherapy, respectively (hazard ratio [HR], 0.91; 95% CI, 0.74 to 1.11; P = .1779). In the PD-L1-positive population (n = 54), the HR for OS was 1.13 (95% CI, 0.57 to 2.23; P = .6352). In an exploratory analysis of the PD-L1-positive population, defined as combined positive score ≥ 1 (22C3 assay; n = 137), median OS was 14.9 months (95% CI, 8.7 to 17.3 months) with avelumab versus 11.6 months (95% CI, 8.4 to 12.6 months) with chemotherapy (unstratified HR, 0.72; 95% CI, 0.49 to 1.05). With avelumab and chemotherapy, treatment-related adverse events (TRAEs) occurred in 149 (61.3%) and 184 (77.3%) patients, including grade ≥ 3 TRAEs in 31 (12.8%) and 78 (32.8%) patients, respectively. CONCLUSION JAVELIN Gastric 100 did not demonstrate superior OS with avelumab maintenance versus continued chemotherapy in patients with advanced GC or GEJC overall or in a prespecified PD-L1-positive population.
Collapse
Affiliation(s)
- Markus Moehler
- Department of Internal Medicine, Johannes-Gutenberg University, Mainz, Germany
| | - Mikhail Dvorkin
- Department of Oncology, Budgetary Healthcare Institution of Omsk Region Clinical Oncology Dispensary, Omsk, Russian Federation
| | | | - Mustafa Özgüroğlu
- Clinical Trial Unit, Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Alina S. Muntean
- Department of Research, Oncology Institute Prof Dr Ion Chiricuţă, Cluj Napoca, Romania
| | - Sara Lonardi
- Dipartimento di Oncologia Clinica e Sperimentale, Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy
| | - Marina Nechaeva
- Arkhangelsk Clinical Oncological Dispensary, State Budgetary Healthcare Institution of Arkhangelsk Region, Arkhangelsk, Russian Federation
| | | | - Hasan S. Coşkun
- Department of Medical Oncology, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Antonio Cubillo Gracian
- Hospital Universitario HM Sanchinarro, Madrid, Spain
- CEU Universidad San Pablo, Madrid, Spain
| | | | - Rachel Wong
- Eastern Health, Box Hill Hospital, Monash University, Melbourne, Victoria, Australia
| | - Howard Safran
- Rhode Island Hospital, Brown University, Providence, RI
| | | | - Zev A. Wainberg
- Ronald Reagan University of California Los Angeles Medical Center, Santa Monica, CA
| | - Matthew R. Silver
- EMD Serono Research & Development Institute, Inc, Billerica, MA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Huiling Xiong
- EMD Serono Research & Development Institute, Inc, Billerica, MA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Janet Hong
- EMD Serono Research & Development Institute, Inc, Billerica, MA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges-Pompidou European Hospital, Assistance Publique–Hôpitaux de Paris, Université de Paris, Paris, France
| | - Yung-Jue Bang
- Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
133
|
Chang SC, Tang CM, Le PH, Kuo CJ, Chen TH, Wang SY, Chou WC, Chen TC, Yeh TS, Hsu JT. Impact of Pancreatic Resection on Survival in Locally Advanced Resectable Gastric Cancer. Cancers (Basel) 2021; 13:1289. [PMID: 33799426 PMCID: PMC8001184 DOI: 10.3390/cancers13061289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 02/04/2023] Open
Abstract
Whether gastric adenocarcinoma (GC) patients with adjacent organ invasion (T4b) benefit from aggressive surgery involving pancreatic resection (PR) remains unclear. This study aimed to clarify the impact of PR on survival in patients with locally advanced resectable GC. Between 1995 and 2017, patients with locally advanced GC undergoing radical-intent gastrectomy with and without PR were enrolled and stratified into four groups: group 1 (G1), pT4b without pancreatic resection (PR); group 2 (G2), pT4b with PR; group 3 (G3), positive duodenal margins without Whipple's operation; and group 4 (G4), cT4b with Whipple's operation. Demographics, clinicopathological features, and outcomes were compared between G1 and G2 and G3 and G4. G2 patients were more likely to have perineural invasion than G1 patients (80.6% vs. 50%, p < 0.001). G4 patients had higher lymph node yield (40.8 vs. 31.3, p = 0.002), lower nodal status (p = 0.029), lower lymph node ratios (0.20 vs. 0.48, p < 0.0001) and higher complication rates (45.2% vs. 26.3%, p = 0.047) than G3 patients. The 5-year disease-free survival (DFS) and overall survival (OS) rates were significantly longer in G1 than in G2 (28.1% vs. 9.3%, p = 0.003; 32% vs. 13%, p = 0.004, respectively). The 5-year survival rates did not differ between G4 and G3 (DFS: 14% vs. 14.4%, p = 0.384; OS: 12.6% vs. 16.4%, p = 0.321, respectively). In conclusion, patients with T4b lesion who underwent PR had poorer survival than those who underwent resection of other adjacent organs. Further Whipple's operation did not improve survival in pT3-pT4 GC with positive duodenal margins.
Collapse
Affiliation(s)
- Shih-Chun Chang
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (S.-C.C.); (C.-M.T.); (S.-Y.W.); (T.-S.Y.)
| | - Chi-Ming Tang
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (S.-C.C.); (C.-M.T.); (S.-Y.W.); (T.-S.Y.)
| | - Puo-Hsien Le
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (P.-H.L.); (C.-J.K.); (T.-H.C.)
| | - Chia-Jung Kuo
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (P.-H.L.); (C.-J.K.); (T.-H.C.)
| | - Tsung-Hsing Chen
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (P.-H.L.); (C.-J.K.); (T.-H.C.)
| | - Shang-Yu Wang
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (S.-C.C.); (C.-M.T.); (S.-Y.W.); (T.-S.Y.)
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan;
| | - Tse-Ching Chen
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan;
| | - Ta-Sen Yeh
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (S.-C.C.); (C.-M.T.); (S.-Y.W.); (T.-S.Y.)
| | - Jun-Te Hsu
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; (S.-C.C.); (C.-M.T.); (S.-Y.W.); (T.-S.Y.)
| |
Collapse
|
134
|
Twomey JD, Zhang B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J 2021; 23:39. [PMID: 33677681 PMCID: PMC7937597 DOI: 10.1208/s12248-021-00574-0] [Citation(s) in RCA: 415] [Impact Index Per Article: 103.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are considered a new standard-of-care across many cancer indications. This review provides an update on ICIs approved by the Food and Drug Administration (FDA), with focus on monoclonal antibodies that target the programmed cell death 1 (PD-1) or its ligand, PD-1 ligand 1 (PD-L1), including information on their clinical indications and associated companion diagnostics. The information is further discussed with strategies for identifying predictive biomarkers to guide the clinical use of PD-1/PD-L1-targeted therapies.
Collapse
Affiliation(s)
- Julianne D Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| |
Collapse
|
135
|
Li X, Huang Q, Lei Y, Zheng X, Dai S, Leng W, Liu M. Locally advanced gastroesophageal junction cancer with pathological complete response to neoadjuvant therapy: a case report and literature review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:513. [PMID: 33850910 PMCID: PMC8039689 DOI: 10.21037/atm-21-434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Most gastric cancer and gastroesophageal junction carcinoma (GEJ) patients are already in the advanced stage at the time of diagnosis. Thus, the probability of radical gastrectomy is low, and surgical treatment alone has a poor prognosis due to the high recurrence rate. In order to reduce the recurrence and distant metastasis after surgery, there have been many attempts made to improve the perioperative treatment of advanced localized gastric cancer, but no uniform criteria exist. Over recent years, immunotherapy has revolutionized cancer treatment, and immune checkpoint inhibitors (ICIs) have shown excellent efficacy across various types of tumors, becoming a potential treatment after surgery, chemotherapy, radiotherapy, and targeted therapy. However, the efficacy of single-agent ICIs for gastric cancer is still unsatisfactory. As comprehensive, chemotherapy-based treatment has become the standard care for locally advanced gastric cancer, exploring combination treatment with immune checkpoint inhibitors (ICIs) may be valuable to improving survival outcomes. Here, we report a 66-year-old male with dysphagia diagnosed with GEJ and was defined as clinical stage (cT4N2M0) and Siewert type II, characterized as mismatch repair proficient (pMMR) and programmed cell death ligand-1 (PD-L1) negative; surprisingly, with anti-PD-1 antibody plus SOX (S-1: a combination of tegafur, gimeracil, and oteracil+ oxaliplatin) as perioperative therapy, the patient achieved pathological complete remission (pCR), which indicates that the addition of ICIs to chemotherapy as a perioperative comprehensive treatment might provide a promising strategy option for GEJ. In addition, we review the current status of perioperative comprehensive treatment, in hope that this may provide some reference value for clinical decision-making.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Huang
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yanna Lei
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiufeng Zheng
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuang Dai
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Weibing Leng
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Liu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
136
|
Wang Z, Kang W, Li O, Qi F, Wang J, You Y, He P, Suo Z, Zheng Y, Liu HM. Abrogation of USP7 is an alternative strategy to downregulate PD-L1 and sensitize gastric cancer cells to T cells killing. Acta Pharm Sin B 2021; 11:694-707. [PMID: 33777676 PMCID: PMC7982505 DOI: 10.1016/j.apsb.2020.11.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Targeting immune checkpoints such as programmed cell death protein 1 (PD-1) and programmed death ligand-1 (PD-L1) have been approved for treating melanoma, gastric cancer (GC) and bladder cancer with clinical benefit. Nevertheless, many patients failed to respond to anti-PD-1/PD-L1 treatment, so it is necessary to seek an alternative strategy for traditional PD-1/PD-L1 targeting immunotherapy. Here with the data from The Cancer Genome Atlas (TCGA) and our in-house tissue library, PD-L1 expression was found to be positively correlated with the expression of ubiquitin-specific processing protease 7 (USP7) in GC. Furthermore, USP7 directly interacted with PD-L1 in order to stabilize it, while abrogation of USP7 attenuated PD-L1/PD-1 interaction and sensitized cancer cells to T cell killing in vitro and in vivo. Besides, USP7 inhibitor suppressed GC cells proliferation by stabilizing P53 in vitro and in vivo. Collectively, our findings indicate that in addition to inhibiting cancer cells proliferation, USP7 inhibitor can also downregulate PD-L1 expression to enhance anti-tumor immune response simultaneously. Hence, these data posit USP7 inhibitor as an anti-proliferation agent as well as a novel therapeutic agent in PD-L1/PD-1 blockade strategy that can promote the immune response of the tumor.
Collapse
Key Words
- BCA, bicinchoninic acid
- CHX, cycloheximide
- CSN5, COP9 signalosome 5
- Cancer biology
- DUB, deubiquitinating enzymes
- EBNA1, Epstein–Barr nuclear antigen 1
- Epigenetics
- FDA, U.S. Food and Drug Administration
- FOXO4, forkhead box O4
- GC, gastric cancer
- GEPIA, Gene-Expression Profiling Interactive Analysis
- Gastric cancer
- H2O2, hydrogen peroxidase
- HAUSP, herpes virus-associated ubiquitin-specific protease
- HDN, well differentiated matched adjacent normal tissues
- HDT, well differentiated tumor tissues
- ICP0, infected cell protein 0
- IL-2, interleukin 2
- Immunosuppression
- Immunotherapy
- MDM2, murine double minute-2
- PBMC, peripheral blood mononuclear cells
- PBS, phosphate buffer saline
- PD-1, programmed cell death protein 1
- PD-L1
- PD-L1, programmed death ligand-1
- PDN, poor differentiated matched adjacent normal tissues
- PDT, poor differentiated tumor tissues
- PTMs, post-translational modifications
- RIPA, radioimmunoprecipitation
- TCGA, the Cancer Genome Atlas
- TCR, T cell receptor
- TILs, tumor-infiltrating T cells
- USP18, ubiquitin specific peptidase 18
- USP22, ubiquitin specific peptidase 22
- USP38, ubiquitin specific peptidase 38
- USP7
- USP7, ubiquitin-specific processing protease 7
- USP9X, ubiquitin specific peptidase 9 X-linked
- Ubiquitination
- WB, Western blotting
- irAEs, immune-related adverse effects
- qRT-PCR, quantitative real time polymerase chain reaction
Collapse
|
137
|
Catanese S, Lordick F. Targeted and immunotherapy in the era of personalised gastric cancer treatment. Best Pract Res Clin Gastroenterol 2021; 50-51:101738. [PMID: 33975679 DOI: 10.1016/j.bpg.2021.101738] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Gastric cancer is a major cause of cancer-related morbidity and mortality worldwide. Advances in targeted medical treatment were scarce in the past and challenged by the marked spatial and temporal biological heterogeneity of gastric cancer. Recent molecular profiling studies have increased our understanding of genetic and epigenetic drivers, leading to better patient selection for drug development. Beyond that, immune-related biomarkers were identified, paving the way for future effective immunotherapy. We systematically reviewed articles from PubMed of the past 10 years, and abstracts from annual meetings of ESMO, ASCO and AACR to summarise the current knowledge about targeted and immunotherapy and outline pathways to future personalised therapy of gastric cancer.
Collapse
Affiliation(s)
- Silvia Catanese
- Unit of Medical Oncology, Department of Oncology, University of Pisa, Pisa, Italy
| | - Florian Lordick
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Centre Leipzig (UCCL), Leipzig University Medical Centre, Leipzig, Germany.
| |
Collapse
|
138
|
Chung HC, Bang YJ, S Fuchs C, Qin SK, Satoh T, Shitara K, Tabernero J, van Cutsem E, Alsina M, Cao ZA, Lu J, Bhagia P, Shih CS, Janjigian YY. First-line pembrolizumab/placebo plus trastuzumab and chemotherapy in HER2-positive advanced gastric cancer: KEYNOTE-811. Future Oncol 2021; 17:491-501. [PMID: 33167735 PMCID: PMC8411394 DOI: 10.2217/fon-2020-0737] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
Treatment options for patients with HER2-positive advanced gastric cancer are limited, and the prognosis for these patients is poor. Pembrolizumab has demonstrated promising antitumor activity in patients with advanced gastric or gastroesophageal junction adenocarcinoma as monotherapy, in combination with chemotherapy and in combination with trastuzumab. Combining pembrolizumab with trastuzumab and chemotherapy may therefore provide a benefit for patients with advanced HER2-positive gastric cancer. Here we aimed to describe the design of and rationale for the randomized, double-blind, placebo-controlled Phase III KEYNOTE-811 study, which will evaluate the efficacy and safety of pembrolizumab or placebo in combination with trastuzumab and chemotherapy as first-line treatment for patients with advanced HER2-positive gastric or gastroesophageal junction adenocarcinoma. Clinical trial registration: NCT03615326 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Hyun Cheol Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT 06510, USA
| | - Shu-Kui Qin
- Cancer Center of People's Liberation Army, Nanjing, 210002, China
| | - Taroh Satoh
- Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kohei Shitara
- National Cancer Center Hospital East, Kashiwa, 277-8577, Japan
| | - Josep Tabernero
- Vall d'Hebron University Hospital & Institute of Oncology, Barcelona, 08035, Spain
| | - Eric van Cutsem
- University Hospitals Gasthuisberg Leuven & KU Leuven, Leuven, 03001, Belgium
| | - Maria Alsina
- Vall d'Hebron University Hospital & Institute of Oncology, Barcelona, 08035, Spain
| | - Zhu Alexander Cao
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Jia Lu
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Pooja Bhagia
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Chie-Schin Shih
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | | |
Collapse
|
139
|
Pan S, Li K, Huang B, Huang J, Xu H, Zhu Z. Efficacy and safety of immune checkpoint inhibitors in gastric cancer: a network meta-analysis of well-designed randomized controlled trials. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:290. [PMID: 33708917 PMCID: PMC7944325 DOI: 10.21037/atm-20-6639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/27/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) that inhibit the programmed death 1 (PD-1)/programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) interactions have shown promising prospects as treatment options for advanced gastric cancer (AGC). This manuscript analyzed well designed clinical trials to evaluate the efficacy and safety of immunotherapy in AGC. METHODS PubMed, Embase, the Cochrane Library, and Medline were searched for randomized controlled trials (RCTs) of AGC treatments that were published before April 2020. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and treatment-related adverse events (TRAEs) were evaluated to determine the efficacy and safety of ICIs. Network meta-analysis was performed using a random-effects model under the Bayesian framework. The ability of each treatment was ranked using the surface under the cumulative ranking (SUCRA) curve. RESULTS Our analysis included five studies having seven immunotherapy regimens and 1,730 patients. The network meta-analysis showed that nivolumab 1 mg/kg every 3 weeks plus ipilimumab 3 mg/kg every 3 weeks (88.369%) was the regimen most likely to improve PFS. Nivolumab 3 mg/kg every 3 weeks (84.563%) and nivolumab 1 mg/kg every 3 weeks plus ipilimumab 3 mg/kg every 3 weeks (84.556%) were similarly best for OS outcome with excellent tolerance. The regimen of avelumab 10 mg/kg every 2 weeks (91.167%) had the lowest TRAEs. All immunotherapies had similar response rates. CONCLUSIONS We recommend nivolumab 3 mg/kg every 2 weeks or nivolumab 1 mg/kg every 3 weeks plus ipilimumab 3 mg/kg every 3 weeks as the preferred regimen due to their high efficacies.
Collapse
Affiliation(s)
- Siwei Pan
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Kai Li
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Baojun Huang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinyu Huang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Huimian Xu
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhi Zhu
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
140
|
Tian C, Jing H, Wang C, Wang W, Cui Y, Chen J, Sha D. Prognostic role of tumour-infiltrating lymphocytes assessed by H&E-stained section in gastric cancer: a systematic review and meta-analysis. BMJ Open 2021; 11:e044163. [PMID: 33518526 PMCID: PMC7853025 DOI: 10.1136/bmjopen-2020-044163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Some studies have identified tumour-infiltrating lymphocytes (TILs) in H&E-stained sections of gastric cancer, but the prognostic and clinicopathological significance of this remains unclear. The objective of this study is to evaluate the associations between H&E-based TIL density and prognosis and clinicopathological characteristics of patients with gastric cancer. DESIGN Systematic review and meta-analysis. DATA SOURCES Cochrane Library, PubMed and Embase databases were searched through 25 February 2020. ELIGIBILITY CRITERIA Studies evaluating the correlations between TILs assessed by H&E-stained sections and prognosis and clinicopathological characteristics of gastric cancer were included. DATA EXTRACTION AND SYNTHESIS Relevant data were extracted and risks of bias were assessed independently by two reviewers. HR and relative risk (RR) with 95% CI were pooled by random-effect models to estimate the associations between TIL density and overall survival (OS) and clinicopathological characteristics, respectively. RESULTS We enrolled nine studies including 2835 cases for the present meta-analysis. High TILs were associated with superior OS (HR=0.68, 95% CI 0.52 to 0.87, p=0.003) compared with low TILs. High TILs were significantly associated with lower depth of invasion (T3-T4 vs T1-T2) (RR=0.58, 95% CI 0.50 to 0.66, p<0.001), less lymph node involvement (presence vs absence) (RR=0.68, 95% CI 0.56 to 0.81, p<0.001) and earlier TNM (tumour, node, metastasis) stage (III-IV vs I-II) (RR=0.68, 95% CI 0.55 to 0.83, p<0.001). TIL density was not associated with age, gender, Lauren classification or histological grade. The methodology for evaluating TIL and its cut-off value varied across different studies, which might affect the results of our meta-analysis. CONCLUSIONS Our meta-analysis suggests that H&E-based TIL density is a reliable biomarker to predict the clinical outcomes of patients with gastric cancer. Multicentre, prospective studies are needed to further confirm our findings. PROSPERO REGISTRATION NUMBER CRD42020169877.
Collapse
Affiliation(s)
- Chunfang Tian
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haiyan Jing
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Caixia Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Weibo Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yangang Cui
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jianpeng Chen
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Dan Sha
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
141
|
van den Ende T, de Clercq NC, van Berge Henegouwen MI, Gisbertz SS, Geijsen ED, Verhoeven RHA, Meijer SL, Schokker S, Dings MPG, Bergman JJGHM, Haj Mohammad N, Ruurda JP, van Hillegersberg R, Mook S, Nieuwdorp M, de Gruijl TD, Soeratram TTD, Ylstra B, van Grieken NCT, Bijlsma MF, Hulshof MCCM, van Laarhoven HWM. Neoadjuvant Chemoradiotherapy Combined with Atezolizumab for Resectable Esophageal Adenocarcinoma: A Single-arm Phase II Feasibility Trial (PERFECT). Clin Cancer Res 2021; 27:3351-3359. [PMID: 33504550 DOI: 10.1158/1078-0432.ccr-20-4443] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/03/2021] [Accepted: 01/22/2021] [Indexed: 01/01/2023]
Abstract
PURPOSE The CROSS trial established neoadjuvant chemoradiotherapy (nCRT) for patients with resectable esophageal adenocarcinoma (rEAC). In the PERFECT trial, we investigated the feasibility and efficacy of nCRT combined with programmed-death ligand-1 (PD-L1) inhibition for rEAC. PATIENTS AND METHODS Patients with rEAC received nCRT according to the CROSS regimen combined with five cycles of atezolizumab (1,200 mg). The primary endpoint was the feasibility of administering five cycles of atezolizumab in ≥75% patients. A propensity score-matched nCRT cohort was used to compare pathologic response, overall survival, and progression-free survival. Exploratory biomarker analysis was performed on repeated tumor biopsies. RESULTS We enrolled 40 patients of whom 85% received all cycles of atezolizumab. Immune-related adverse events of any grade were observed in 6 patients. In total, 83% proceeded to surgery. Reasons for not undergoing surgery were progression (n = 4), patient choice (n = 2), and death (n = 1). The pathologic complete response rate was 25% (10/40). No statistically significant difference in response or survival was found between the PERFECT and the nCRT cohort. Baseline expression of an established IFNγ signature was higher in responders compared with nonresponders (P = 0.043). On-treatment nonresponders showed either a high number of cytotoxic lymphocytes (CTL) with a transcriptional signature consistent with expression of immune checkpoints, or a low number of CTLs. CONCLUSIONS Combining nCRT with atezolizumab is feasible in patients with rEAC. On the basis of our exploratory biomarker study, future studies are necessary to elucidate the potential of neoadjuvant immunotherapy in patient subgroups.See related commentary by Catenacci, p. 3269.
Collapse
Affiliation(s)
- Tom van den Ende
- Amsterdam UMC, Department of Medical Oncology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands.
| | - Nicolien C de Clercq
- Amsterdam UMC, Department of Internal and Vascular Medicine, University of Amsterdam, Amsterdam, the Netherlands
| | - Mark I van Berge Henegouwen
- Amsterdam UMC, Department of Surgery, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Suzanne S Gisbertz
- Amsterdam UMC, Department of Surgery, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - E D Geijsen
- Amsterdam UMC, Department of Radiotherapy, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - R H A Verhoeven
- Amsterdam UMC, Department of Medical Oncology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands.,Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Sybren L Meijer
- Amsterdam UMC, Department of Pathology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Sandor Schokker
- Amsterdam UMC, Department of Medical Oncology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - M P G Dings
- Amsterdam UMC, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Jacques J G H M Bergman
- Amsterdam UMC, Department of Gastroenterology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Nadia Haj Mohammad
- Department of Medical Oncology, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jelle P Ruurda
- Department of Surgery, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Stella Mook
- Department of Radiotherapy, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Max Nieuwdorp
- Amsterdam UMC, Department of Internal and Vascular Medicine, University of Amsterdam, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Amsterdam UMC, Department of Medical Oncology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tanya T D Soeratram
- Amsterdam UMC, Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Amsterdam UMC, Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Nicole C T van Grieken
- Amsterdam UMC, Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Maarten C C M Hulshof
- Amsterdam UMC, Department of Radiotherapy, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - H W M van Laarhoven
- Amsterdam UMC, Department of Medical Oncology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
142
|
Xu J, Xu N, Bai Y, Liu R, Mao C, Sui H, Wang X, Jiang Q, Dou Y. Anti-PD-1 antibody HX008 combined with oxaliplatin plus capecitabine for advanced gastric or esophagogastric junction cancer: a multicenter, single-arm, open-label, phase Ib trial. Oncoimmunology 2020; 10:1864908. [PMID: 33457083 PMCID: PMC7781732 DOI: 10.1080/2162402x.2020.1864908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Anti-PD-1 monoclonal antibody is approved as an option for third-line treatment of advanced gastric and gastroesophageal junction (G/GEJ) cancer in several countries, but no anti-PD-1 monoclonal antibody treatment is yet approved for first-line treatment of advanced G/GEJ cancer. We report a phase Ib trial of HX008, a highly selective, humanized anti-programmed death-1 monoclonal antibody, plus oxaliplatin and capecitabine as first-line treatment for advanced G/GEJ cancer. Patients with previously untreated, locally advanced or metastatic G/GEJ cancer were enrolled. All patients received HX008 3 mg/kg intravenously every 3 weeks, oxaliplatin 130 mg/m2 intravenously on day 1 every 3 weeks (up to 6 cycles), and capecitabine 1000 mg/m2 orally twice daily for 14 days continuous dosing followed by a 7-day break. The primary end point was the incidence of adverse events and serious adverse events. In total, 35 patients were enrolled. Median follow-up was 12.7 months. Most frequent (>10%) grade ≥3 treatment-related adverse events were anemia (27.5%), neutropenia (20%), thrombocytopenia (17.1%), leukopenia (17.1%) and fatigue (17.3%). Objective response rate was 60.0% (95% confidence interval [CI] 42.1-76.1%). Disease control rate was 77.1% (95% CI 59.9-89.6). Median time to response and duration of response were 1.4 months (range 1.3-2.9) and 12.3 months (range 1.4-17.9+), respectively. Median PFS was 9.2 months (95% CI 5.4-not reached). These results demonstrated that HX008 combined with oxaliplatin plus capecitabine was well tolerated and demonstrated encouraging efficacy as first-line treatment for advanced G/GEJ cancer. This study was registered in china, register number was CTR20181270.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Nong Xu
- Department of Medical Oncology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yuxian Bai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Rongrui Liu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chenyu Mao
- Department of Medical Oncology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Hong Sui
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaofei Wang
- Taizhou Hanzhong Biomedical Co., Ltd., Jiangsu, China
| | - Qian Jiang
- Taizhou Hanzhong Biomedical Co., Ltd., Jiangsu, China
| | - Yiwei Dou
- Taizhou Hanzhong Biomedical Co., Ltd., Jiangsu, China
| |
Collapse
|
143
|
Paver EC, Cooper WA, Colebatch AJ, Ferguson PM, Hill SK, Lum T, Shin JS, O'Toole S, Anderson L, Scolyer RA, Gupta R. Programmed death ligand-1 (PD-L1) as a predictive marker for immunotherapy in solid tumours: a guide to immunohistochemistry implementation and interpretation. Pathology 2020; 53:141-156. [PMID: 33388161 DOI: 10.1016/j.pathol.2020.10.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022]
Abstract
Immunotherapy with checkpoint inhibitors is well established as an effective treatment for non-small cell lung cancer and melanoma. The list of approved indications for treatment with PD-1/PD-L1 checkpoint inhibitors is growing rapidly as clinical trials continue to show their efficacy in patients with a wide range of solid tumours. Clinical trials have used a variety of PD-L1 immunohistochemical assays to evaluate PD-L1 expression on tumour cells, immune cells or both as a potential biomarker to predict response to immunotherapy. Requests to pathologists for PD-L1 testing to guide choice of therapy are rapidly becoming commonplace. Thus, pathologists need to be aware of the different PD-L1 assays, methods of evaluation in different tumour types and the impact of the results on therapeutic decisions. This review discusses the key practical issues relating to the implementation of PD-L1 testing for solid tumours in a pathology laboratory, including evidence for PD-L1 testing, different assay types, the potential interchangeability of PD-L1 antibody clones and staining platforms, scoring criteria for PD-L1, validation, quality assurance, and pitfalls in PD-L1 assessment. This review also explores PD-L1 IHC in solid tumours including non-small cell lung carcinoma, head and neck carcinoma, triple negative breast carcinoma, melanoma, renal cell carcinoma, urothelial carcinoma, gastric and gastroesophageal carcinoma, colorectal carcinoma, hepatocellular carcinoma, and endometrial carcinoma. The review aims to provide pathologists with a practical guide to the implementation and interpretation of PD-L1 testing by immunohistochemistry.
Collapse
Affiliation(s)
- Elizabeth C Paver
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Wendy A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Andrew J Colebatch
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Peter M Ferguson
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Sean K Hill
- Gold Coast University Hospital, Southport, Qld, Australia
| | - Trina Lum
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Joo-Shik Shin
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Sandra O'Toole
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Lyndal Anderson
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Richard A Scolyer
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Ruta Gupta
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
144
|
Xie J, Fu L, Jin L. Immunotherapy of gastric cancer: Past, future perspective and challenges. Pathol Res Pract 2020; 218:153322. [PMID: 33422778 DOI: 10.1016/j.prp.2020.153322] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/08/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022]
Abstract
Gastric cancer is considered as the third leading cause of deaths and the fifth most common cancers worldwide. Common treatment approaches include chemotherapy, radiation, gastric resection and targeted therapies. The emergence of gastric cancer immunotherapy has already shown some promising results and have altered the therapeutic procedures. Now, different combination therapies as well as novel immunotherapies targeting new molecules have been proposed. Despite ongoing investigations on the therapeutic options and significant advancements in this regard, the disease is poorly prognosed. In fact, limited therapeutic options and delayed diagnosis lead to the progression, dissemination and metastasis of the disease. Current immunotherapies are mostly based on cytotoxic immunocytes, monoclonal antibodies and gene transferred vaccines. The use of Immune checkpoint inhibitors (ICIs) have grown rapidly. In this review, we aimed to explore perspective and progression of different approaches of immunotherapy in the treatment of GC and the clinical outcomes reported so far. We also summarized the tumor immunosurveillance and tumor immunoescape.
Collapse
Affiliation(s)
- Jun Xie
- Department of Gastroenterology Surgery, Affiliated Hospital of Shaoxing University, Shaoxing 312000, Zhejiang Province, China
| | - Liping Fu
- Department of Nuclear Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Li Jin
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
145
|
Ng MCH, Choo SP. Combination of immunotherapy with chemotherapy in first line treatment of metastatic gastric cancer? Too much, too little or just right? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1692. [PMID: 33490204 PMCID: PMC7812240 DOI: 10.21037/atm-20-4699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Matthew C. H. Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | |
Collapse
|
146
|
Ramos MFKP, Pereira MA, de Castria TB, Ribeiro RRE, Cardili L, de Mello ES, Zilberstein B, Ribeiro-Júnior U, Cecconello I. Remnant gastric cancer: a neglected group with high potential for immunotherapy. J Cancer Res Clin Oncol 2020; 146:3373-3383. [PMID: 32671505 DOI: 10.1007/s00432-020-03322-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/10/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE The importance of targeted therapy and interest in the study of predictive markers in gastric cancer (GC) have increased in recent years with the use of anti-HER2 therapy and immunotherapy with anti-PD1/PD-L1 for microsatellite instability (MSI) and PD-L1 + tumors. However, the behavior of remnant GC (RGC) in this scenario is poorly reported. Thus, this study aims to evaluate the clinicopathological characteristics and prognosis of RGC and its association with the expression of current markers for targeted therapy. METHODS All RGC resections performed in a single center from 2009 to 2019 were retrospectively reviewed. As a comparison group, 53 primary proximal GC (PGC) who underwent total D2-gastrectomy were selected. HER2, MSI status and PD-L1 expression were analyzed by immunohistochemistry. Combined Positive Score (CPS) was used to determine PD-L1 positivity. RESULTS A total of 40 RGC were included. RGC patients were older (p = 0.001), had lower BMI (p = 0.001) and number of resected lymph nodes (p < 0.001) compared to the PGC. Regarding markers expression, MSI was higher in RGC than PGC (27.5% vs 9.4%, p = 0.022). The frequency of CPS-positive was 32.5% and 26.4% in RGC and PGC, respectively (p = 0.522). HER2 positivity was 17.5% and 22.6% for RGC and PGC, respectively (p = 0.543). In survival analysis, DFS was better for RGC CPS-positive than RGC CPS-negative (p = 0.039) patients. There was no difference in survival considering MSI status. CONCLUSION RGC had higher incidence of MSI than PGC, and CPS-positive RGC was associated with better survival. The immunological profile of RGC patients suggests that they would be good candidates for immunotherapy.
Collapse
Affiliation(s)
- Marcus Fernando Kodama Pertille Ramos
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil.
| | - Marina Alessandra Pereira
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Tiago Biachi de Castria
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Renan Ribeiro E Ribeiro
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Leonardo Cardili
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Evandro Sobroza de Mello
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Bruno Zilberstein
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Ulysses Ribeiro-Júnior
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| | - Ivan Cecconello
- Instituto do Cancer, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av Dr Arnaldo 251, Sao Paulo, SP, 01249000, Brazil
| |
Collapse
|
147
|
Abstract
In recent years, there have been advancements in traditional patterns of tumor therapy with the adoption of immunotherapy. Its application with or without other combined regimens has attracted attention from clinicians. Sintilimab (Tyvyt®), a highly selective fully human IgG4 monoclonal antibody, blocks the binding site of programmed cell death protein 1 (PD-1), thereby, inhibiting the interaction between PD-1 and its ligands (PD-L1/2) to restore the endogenous anti-tumor T cell responses. Sintilimab has been proven to be clinically beneficial in multiple solid tumor therapies. Combination therapy and monotherapy have shown potential and encouraging anti-tumor efficacy with controllable and acceptable toxicities. The combination therapy is more likely to be a novel and promising therapeutic option. This study provides an overview of the status of sintilimab-based clinical trials in various solid tumors.
Collapse
|
148
|
Yang L, Dong XZ, Xing XX, Cui XH, Li L, Zhang L. Efficacy and safety of anti-PD-1/anti-PD-L1 antibody therapy in treatment of advanced gastric cancer or gastroesophageal junction cancer: A meta-analysis. World J Gastrointest Oncol 2020; 12:1346-1363. [PMID: 33250966 PMCID: PMC7667450 DOI: 10.4251/wjgo.v12.i11.1346] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 09/18/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Faced with limited and inadequate treatment options for patients with advanced gastric cancer or gastroesophageal junction cancer (GC/GEJC), researchers have turned toward, with the support of promising clinical trials, anti-PD-1/anti-PD-L1 antibody therapy. But there are also different clinical trial results. To better assess its efficacy and safety, we integrated data from 13 eligible studies for a systematic review and meta-analysis.
AIM To comprehensively evaluate the efficacy and safety of anti-PD-1/anti-PD-L1 antibody therapy in the treatment of advanced GC/GEJC patients.
METHODS PubMed, Web of Science, Cochrane Library ,and EMBASE databases were searched to identify eligible articles with outcomes including objective response rate (ORR), disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and adverse events (AEs) of anti-PD-1/anti-PD-L1 antibody therapy.
RESULTS Our study encompassed a total of 13 trials totaling 1618 patients. The outcomes showed a pooled ORR and DCR of 15% (95% confidence interval [CI]: 14%-18%) and 40% (95%CI: 33%-46%), respectively. The pooled 6-mo OS and PFS were 54% (95%CI: 45%-64%) and 26% (95%CI: 20%-32%), respectively, and the 12-mo OS and PFS were 42% (95%CI: 21%-62%) and 11% (95%CI: 8%-13%), respectively. In addition, the incidence of any-grade AEs and grade ≥ 3 AEs was 64% (95%CI: 54%-73%) and 18% (95%CI: 16%-20%), respectively. Most importantly, PD-L1 positive patients exhibited a higher ORR rate than PD-L1 negative patients (odds ratio = 2.54, 95%CI: 1.56-4.15).
CONCLUSION Anti-PD-1/anti-PD-L1 antibody therapy has shown promising anti-tumor efficacy with manageable AEs in advanced GC/GEJC patients, with PD-L1 overexpressing patients exhibiting a higher ORR. What is more, the clinical efficacy of anti-PD-1/PD-L1 combined with traditional chemotherapy drugs is even better, although the occurrence of AEs still causes considerate concerns.
Collapse
Affiliation(s)
- Li Yang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Xian-Zhe Dong
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Xiao-Xuan Xing
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Xiao-Hui Cui
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| |
Collapse
|
149
|
Liu C, Wang Y, Li S, Jiao X, Zou J, Wang Z, Qi C, Zhang X, Li J, Lu Z, Shen L. Early change in peripheral CD4 + T cells associated with clinical outcomes of immunotherapy in gastrointestinal cancer. Immunotherapy 2020; 13:55-66. [PMID: 33086925 DOI: 10.2217/imt-2020-0068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Biomarkers for immune checkpoint inhibitors (ICIs) are limited in gastrointestinal cancer. Peripheral blood lymphocyte subset and associated dynamic changes were retrospectively analyzed in patients with gastrointestinal cancer treated with ICIs. Cox regression and Kaplan-Meier analyses were conducted for survival. A total of 80 patients were enrolled. Baseline CD4+/CD8+ T cells were lower in patients who experienced tumor progression by 6 months than in patients who did not (1.160 ± 0.652 vs 1.705 ± 0.924, respectively; p = 0.003). In multivariate analyses, decline in CD4+ T cells after the first dose of ICIs (CD4-C1-decline) was an independent prognostic factor for overall survival (hazard ratio: 13.00; 95% CI: 2.24-75.54; p = 0.004). Furthermore, CD4-C1-decline was a preferable indicator for progression in patients with deficient mismatch repair/microsatellite instability-high (p = 0.027). Early change in CD4+ T cell counts in peripheral blood may act as a prognostic biomarker for gastrointestinal cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Chang Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Yanni Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Shuang Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Xi Jiao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Jianling Zou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China
| |
Collapse
|
150
|
Ramucirumab in Combination with Pembrolizumab in Treatment-Naïve Advanced Gastric or GEJ Adenocarcinoma: Safety and Antitumor Activity from the Phase 1a/b JVDF Trial. Cancers (Basel) 2020; 12:cancers12102985. [PMID: 33076423 PMCID: PMC7602637 DOI: 10.3390/cancers12102985] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The prognosis for gastric cancer remains poor, with a median overall survival of approximately 1 year. Ramucirumab and pembrolizumab have each demonstrated antitumor activity and a favorable safety profile as treatments for patients with advanced gastric/gastroesophageal junction (G/GEJ) cancer in the second and third-line setting respectively. However, both agents failed to demonstrate survival benefit over chemotherapy in the first-line setting. Twenty-eight treatment-naïve patients with advanced/metastatic G/GEJ adenocarcinoma were treated with ramucirumab plus pembrolizumab in this phase 1a/b trial. Our results showed that this combination was well tolerated with no unexpected toxicities, and promising durable survival results, particularly among patients with PD-ligand 1 positive tumors. The results of our study therefore support modulating the tumor microenvironment with dual inhibition of VEGFR2 and PD-1 pathways in the first-line patients with advanced G/GEJ cancer. Abstract Ramucirumab (anti-VEGFR2) plus pembrolizumab (anti-PD1) demonstrated promising antitumor activity and tolerability among patients with previously treated advanced cancers, supporting growing evidence that combination therapies modulating the tumor microenvironment may expand the spectrum of patients who respond to checkpoint inhibitors. Here we present the results of this combination in first-line patients with metastatic G/GEJ cancer. Twenty-eight patients (≥18 years) with no prior systemic chemotherapy in the advanced/metastatic setting received ramucirumab (8 mg/kg days 1 and 8) plus pembrolizumab (200 mg day 1) every 3 weeks as part of JVDF phase 1a/b study. The primary endpoint was safety. Secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and overall survival (OS). Tumors were PD-L1-positive (combined positive score ≥ 1) in 19 and -negative in 6 patients. Eighteen patients experienced grade 3 treatment-related adverse events, most commonly hypertension (14%) and elevated alanine/aspartate aminotransferase (11% each), with no grade 4 or 5 reported. The ORR was 25% (PD-L1-positive, 32%; PD-L1-negative, 17%) with duration of response not reached. PFS was 5.6 months (PD-L1-positive, 8.6 months; PD-L1-negative, 4.3 months), and OS 14.6 months (PD-L1-positive, 17.3 months; PD-L1-negative, 11.3 months). Acknowledging study design limitations, ramucirumab plus pembrolizumab had encouraging durable clinical activity with no unexpected toxicities in treatment-naïve biomarker-unselected metastatic G/GEJ cancer, and improved outcomes in patients with PD-L1-positive tumors.
Collapse
|