801
|
The effect of adding immune checkpoint inhibitors on the risk of pneumonitis for solid tumours: a meta-analysis of phase III randomised controlled trials. Eur J Cancer 2021; 150:168-178. [PMID: 33906099 DOI: 10.1016/j.ejca.2021.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/20/2021] [Accepted: 03/04/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have become one of the standard treatment options for solid tumours; however, treatment-related adverse events, including pneumonitis, sometimes disrupt the ongoing treatment. To evaluate the impact of ICI addition on the risk of pneumonitis, a meta-analysis was conducted. METHODS Phase III randomised controlled trials comparing ICIs and conventional therapy with conventional therapy alone were selected by searching databases, including PubMed, Embase, Web of Science and Cochrane Library. The odds ratio (OR) of any grade and grade III-V pneumonitis was calculated. Meta-analysis was performed to compare the incidence of pneumonitis between the treatment arm with additional ICIs and the arm with conventional therapy. RESULTS A total of 25 randomised controlled trials (RCTs) representing 16,343 patients for grade I-V and 23 RCTs with 15,006 patients for grade III-V pneumonitis were analysed. Adding ICIs was associated with a significant increase in pneumonitis (grade I-V: OR, 2.67, 95% confidence interval [CI]: 2.12-3.37, grade III-V: OR, 1.83, 95% CI: 1.26-2.65). In subgroup analyses based on the mechanism of action of ICIs, cancer types (lung and non-lung cancer) and each ICI, no significant difference in heterogeneity was observed. CONCLUSIONS Adding ICIs to the conventional treatment for solid tumours significantly increased both grade I-V and grade III-V pneumonitis regardless of the mechanisms of ICIs and cancer type. These results would be helpful for oncologists to choose the appropriate treatment options using ICIs, particularly for patients with known risk factors of pneumonitis or interstitial lung disease.
Collapse
|
802
|
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
|
803
|
Rimini M, Pecchi A, Prampolini F, Bussei C, Salati M, Forni D, Martelli F, Valoriani F, Canino F, Bocconi A, Gelsomino F, Reverberi L, Benatti S, Piacentini F, Menozzi R, Dominici M, Luppi G, Spallanzani A. The Prognostic Role of Early Skeletal Muscle Mass Depletion in Multimodality Management of Patients with Advanced Gastric Cancer Treated with First Line Chemotherapy: A Pilot Experience from Modena Cancer Center. J Clin Med 2021; 10:jcm10081705. [PMID: 33921004 PMCID: PMC8071389 DOI: 10.3390/jcm10081705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Few data about the link between nutritional status and survival are available in the metastatic gastric cancer (GC) setting. The aim of this work was to evaluate the prognostic role of tissue modifications during treatment and the benefit of a scheduled nutritional assessment in this setting. Methods: Clinical and laboratory variables of 40 metastatic GC patients treated at Modena Cancer Center were retrieved: 20 received a nutritional assessment on the oncology’s discretion, the other 20 received a scheduled nutritional assessment at baseline and every 2–4 weeks. Anthropometric parameters were calculated on Computed Tomography (CT) images at the baseline and after 3 months of chemotherapy. Results: A correlation between baseline Eastern Cooperative Oncology Group Performance Status (ECOG PS), Lymphocyte to Monocyte Ratio (LMR), C-reactive protein (PCR), Prognostic Nutritional Index (PNI) and Overall survival (OS) was highlighted. Among the anthropometric parameters, early skeletal muscle mass depletion (ESMMD) >10% in the first months of treatment significantly impacted on mOS (p = 0.0023). A link between ESMMD and baseline LDH > 460 U/L, baseline CRP > 2.2 mg/dL and weight decrease during treatment emerged. Patients evaluated with a nutritional scheduled support experienced a mean gain in subcutaneous and visceral fat of 11.4% and 10.21%, respectively. Conclusion: We confirm the prognostic impact of ESMMD > 10% during chemotherapy in metastatic GC. The prognostic role of a scheduled nutritional assessment deserves further confirmation in large prospective trials.
Collapse
Affiliation(s)
- Margherita Rimini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Annarita Pecchi
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Francesco Prampolini
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Chiara Bussei
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Daniela Forni
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Francesca Martelli
- Department of Radiology, University Hospital of Modena, 41122 Modena, Italy; (A.P.); (F.P.); (D.F.); (F.M.)
| | - Filippo Valoriani
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Fabio Canino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Alessandro Bocconi
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Fabio Gelsomino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Linda Reverberi
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Stefania Benatti
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Federico Piacentini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Renata Menozzi
- Division of Metabolic Disease and Clinical Nutrition, University Hospital of Modena, 41122 Modena, Italy; (C.B.); (F.V.); (L.R.); (R.M.)
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Gabriele Luppi
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
| | - Andrea Spallanzani
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41122 Modena, Italy; (M.R.); (M.S.); (F.C.); (A.B.); (F.G.); (S.B.); (F.P.); (M.D.); (G.L.)
- Correspondence: ; Tel.: +39-05-9422-3310
| |
Collapse
|
804
|
Kwon M, An M, Klempner SJ, Lee H, Kim KM, Sa JK, Cho HJ, Hong JY, Lee T, Min YW, Kim TJ, Min BH, Park WY, Kang WK, Kim KT, Kim ST, Lee J. Determinants of Response and Intrinsic Resistance to PD-1 Blockade in Microsatellite Instability-High Gastric Cancer. Cancer Discov 2021; 11:2168-2185. [PMID: 33846173 DOI: 10.1158/2159-8290.cd-21-0219] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/28/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022]
Abstract
Sequence alterations in microsatellites and an elevated mutational burden are observed in 20% of gastric cancers and associated with clinical response to anti-PD-1 antibodies. However, 50% of microsatellite instability-high (MSI-H) cancers are intrinsically resistant to PD-1 therapies. We conducted a phase II trial of pembrolizumab in patients with advanced MSI-H gastric cancer and included serial and multi-region tissue samples in addition to serial peripheral blood analyses. The number of whole-exome sequencing (WES)-derived nonsynonymous mutations correlated with antitumor activity and prolonged progression-free survival (PFS). Coupling WES to single-cell RNA sequencing, we identified dynamic tumor evolution with greater on-treatment collapse of mutational architecture in responders. Diverse T-cell receptor repertoire was associated with longer PFS to pembrolizumab. In addition, an increase in PD-1+ CD8+ T cells correlated with durable clinical benefit. Our findings highlight the genomic, immunologic, and clinical outcome heterogeneity within MSI-H gastric cancer and may inform development of strategies to enhance responsiveness. SIGNIFICANCE: This study highlights response heterogeneity within MSI-H gastric cancer treated with pembrolizumab monotherapy and underscores the potential for extended baseline and early on-treatment biomarker analyses to identify responders. The observed markers of intrinsic resistance have implications for patient stratification to inform novel combinations among patients with intrinsically resistant features.See related commentary by Fontana and Smyth, p. 2126.This article is highlighted in the In This Issue feature, p. 2113.
Collapse
Affiliation(s)
- Minsuk Kwon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Minae An
- Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul, Korea
| | - Samuel J Klempner
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hyuk Lee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jason K Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Hee Jin Cho
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul, Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Taehyang Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yang Won Min
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Jun Kim
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung-Hoon Min
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyu-Tae Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, Korea.
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. .,Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
805
|
Fundytus A, Booth CM, Tannock IF. How low can you go? PD-L1 expression as a biomarker in trials of cancer immunotherapy. Ann Oncol 2021; 32:833-836. [PMID: 33839260 DOI: 10.1016/j.annonc.2021.03.208] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Affiliation(s)
- A Fundytus
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, Canada; Department of Oncology, Queen's University, Kingston, Canada
| | - C M Booth
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, Canada; Department of Oncology, Queen's University, Kingston, Canada; Department of Public Health Sciences, Queen's University, Kingston, Canada
| | - I F Tannock
- Division of Medical Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, Canada.
| |
Collapse
|
806
|
Park R, Da Silva LL, Saeed A. Immunotherapy Predictive Molecular Markers in Advanced Gastroesophageal Cancer: MSI and Beyond. Cancers (Basel) 2021; 13:1715. [PMID: 33916348 PMCID: PMC8038572 DOI: 10.3390/cancers13071715] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Advanced gastroesophageal cancer (GEC) has a poor prognosis and limited treatment options. Immunotherapy including the anti-programmed death-1 (PD-1) antibodies pembrolizumab and nivolumab have been approved for use in various treatment settings in GEC. Additionally, frontline chemoimmunotherapy regimens have recently demonstrated promising efficacy in large phase III trials and have the potential to be added to the therapeutic armamentarium in the near future. There are currently several immunotherapy biomarkers that are validated for use in the clinical setting for GEC including programmed death ligand-1 (PD-L1) expression as well as the tumor agnostic biomarkers such as mismatch repair or microsatellite instability (MMR/MSI) and tumor mutational burden (TMB). However, apart from MMR/MSI, these biomarkers are imperfect because none are highly sensitive nor specific. Therefore, there is an unmet need for immunotherapy biomarker development. To this end, several biomarkers are currently being evaluated in ongoing trials with some showing promising predictive potential. Here, we summarize the landscape of immunotherapy predictive biomarkers that are currently being evaluated in GEC.
Collapse
Affiliation(s)
- Robin Park
- Department of Medicine, MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA 01702, USA; (R.P.); (L.L.D.S.)
| | - Laercio Lopes Da Silva
- Department of Medicine, MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA 01702, USA; (R.P.); (L.L.D.S.)
| | - Anwaar Saeed
- Department of Medicine, Division of Medical Oncology, Kansas University Medical Center, Kansas City, KS 66205, USA
| |
Collapse
|
807
|
Wang JY, Xiu J, Baca Y, Arai H, Battaglin F, Kawanishi N, Soni S, Zhang W, Millstein J, Shields AF, Grothey A, Weinberg BA, Marshall JL, Lou E, Khushman M, Sohal DPS, Hall MJ, Oberley M, Spetzler D, Shen L, Korn WM, Lenz HJ. Distinct genomic landscapes of gastroesophageal adenocarcinoma depending on PD-L1 expression identify mutations in RAS-MAPK pathway and TP53 as potential predictors of immunotherapy efficacy. Ann Oncol 2021; 32:906-916. [PMID: 33798656 DOI: 10.1016/j.annonc.2021.03.203] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/21/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The impact of molecular alterations on programmed death-ligand 1 (PD-L1) combined positive score (CPS) is not well studied in gastroesophageal adenocarcinomas (GEAs). We aimed to characterize genomic features of tumors with different CPSs in GEAs. PATIENTS AND METHODS Genomic alterations of 2518 GEAs were compared in three groups (PD-L1 CPS ≥ 10, high; CPS = 1-9, intermediate; CPS < 1, low) using next-generation sequencing. We assessed the impact of gene mutations on the efficacy of immune checkpoint inhibitors (ICIs) and tumor immune environment based on the Memorial Sloan Kettering Cancer Center and The Cancer Genome Atlas databases. RESULTS High, intermediate, and low CPSs were seen in 18%, 54% and 28% of GEAs, respectively. PD-L1 positivity was less prevalent in women and in tissues derived from metastatic sites. PD-L1 CPS was positively associated with mismatch repair deficiency/microsatellite instability-high, but independent of tumor mutation burden distribution. Tumors with mutations in KRAS, TP53, and RAS-mitogen-activated protein kinase (MAPK) pathway were associated with higher PD-L1 CPSs in the mismatch repair proficiency and microsatellite stability (pMMR&MSS) subgroup. Patients with RAS-MAPK pathway alterations had longer overall survival (OS) from ICIs compared to wildtype (WT) patients [27 versus 13 months, hazard ratio (HR) = 0.36, 95% confidence interval (CI): 0.19-0.7, P = 0.016] and a similar trend was observed in the MSS subgroup (P = 0.11). In contrast, patients with TP53 mutations had worse OS from ICIs compared to TP53-WT patients in the MSS subgroup (5 versus 21 months, HR = 2.39, 95% CI: 1.24-4.61, P = 0.016). CONCLUSIONS This is the largest study to investigate the distinct genomic landscapes of GEAs with different PD-L1 CPSs. Our data may provide novel insights for patient selection using mutations in TP53 and RAS-MAPK pathway and for the development of rational combination immunotherapies in GEAs.
Collapse
Affiliation(s)
- J Y Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China; Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - J Xiu
- Caris Life Sciences, Phoenix, USA
| | - Y Baca
- Caris Life Sciences, Phoenix, USA
| | - H Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - F Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - N Kawanishi
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - S Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - W Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - J Millstein
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - A F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, USA
| | - A Grothey
- GI Cancer Research, West Cancer Center and Research Institute, Germantown, USA
| | - B A Weinberg
- Division of Hematology and Oncology, Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, USA
| | - J L Marshall
- Division of Hematology and Oncology, Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, USA
| | - E Lou
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| | - M Khushman
- Department of Interdisciplinary Clinical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, USA
| | - D P S Sohal
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, USA
| | - M J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, USA
| | | | | | - L Shen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - W M Korn
- Caris Life Sciences, Phoenix, USA
| | - H J Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
808
|
Wang SW, Sheng H, Zheng F, Zhang F. Hesperetin promotes DOT1L degradation and reduces histone H3K79 methylation to inhibit gastric cancer metastasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153499. [PMID: 33667841 DOI: 10.1016/j.phymed.2021.153499] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/07/2021] [Accepted: 02/04/2021] [Indexed: 05/27/2023]
Abstract
BACKGROUND There have been many researches on the effects of flavonoids on tumor treatment or adjuvant therapy, but there are few studies revealing their epigenetic effect on tumors. Hesperetin is a common citrus flavanone widely distributed among citrus fruits. The role of hesperetin in gastric cancer metastasis is unclear. PURPOSE To investigate the effect of hesperetin on gastric cancer metastasis and its underlying mechanism. METHODS We used cancer cell lines cultured in medium and nude mice implantation as in vitro and in vivo models to investigate the impact of hesperetin treatment on the migration and invasion of gastric cancer cells. The molecular biological experiments such as transwell assay, western blotting, qPCR, ChIP-qPCR, immunostaining and transfection were conducted to explore the molecular mechanisms. RESULTS We found that hesperetin obviously reduced the protein abundance of DOT1L and the methylation of histone H3K79 in a variety of cells. In gastric cancer cells, the treatment of hesperetin decreased cell migration and invasion and the expression of genes closely related to the metastatic capability. Mechanistically, hesperetin affected the stability of DOT1L protein by regulating the activity of CBP. CONCLUSION These findings highlight the epigenetic effect of hesperetin and provide a new perspective to understand the tumor suppressive effect of flavonoids.
Collapse
Affiliation(s)
- Si-Wei Wang
- Quzhou Hospital, Zhejiang University School of Medicine, Quzhou 324000, China; Department of Core Facility, Quzhou People's Hospital, Quzhou 324000, China
| | - Hao Sheng
- Quzhou Hospital, Zhejiang University School of Medicine, Quzhou 324000, China; Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fang Zheng
- Department of Core Facility, Quzhou People's Hospital, Quzhou 324000, China
| | - Feng Zhang
- Quzhou Hospital, Zhejiang University School of Medicine, Quzhou 324000, China; Department of Core Facility, Quzhou People's Hospital, Quzhou 324000, China; Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou 324000, China.
| |
Collapse
|
809
|
Evrard C, Louvet C, Hajbi FE, Fiore FDI, Malicot KLE, Aparicio T, Bouché O, Laurent-Puig P, Bibeau F, Lecomte T, Lièvre A, Guimbaud R, Kim S, Zaanan A, Sokol H, Chibaudel B, Desrame J, Pierre S, Gonzalez D, Lepage C, Tougeron D. PRODIGE 59-DURIGAST trial: A randomised phase II study evaluating FOLFIRI + Durvalumab ± Tremelimumab in second-line of patients with advanced gastric cancer. Dig Liver Dis 2021; 53:420-426. [PMID: 33358124 DOI: 10.1016/j.dld.2020.11.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022]
Abstract
Gastric or gastro-oesophageal junction (GEJ) adenocarcinomas present poor overall survival (OS). First-line chemotherapy regimen for patients with HER2-negative tumours is based on a doublet or triplet of fluoropyrimidine plus platinum salt ± taxane. Second-line chemotherapy (Docetaxel or Irinotecan) improves OS which nonetheless remains poor (around 5 months). The first results of immune checkpoint inhibitors (anti-PD-1) combined with chemotherapy in metastatic gastric and GEJ cancers were discordant in recent phase III trials. Data on dual-blockade (anti-PD-L1 or anti-PD-1 plus anti-CTLA-4) plus chemotherapy are lacking. DURIGAST is a randomised, multicenter, non-comparative, phase II study, evaluating safety and efficacy of FOLFIRI plus Durvalumab (anti-PD-L1) versus FOLFIRI plus Durvalumab and Tremelimumab (anti-CTLA-4) as second-line treatment of advanced gastric and GEJ adenocarcinoma. The primary objective is the rate of patients alive and without progression at 4 months. The main inclusion criteria are: patients with advanced gastric or GEJ adenocarcinoma, pre-treated with fluoropyrimidine + platinum salt ± taxane. Due to a lack of data on FOLFIRI, Durvalumab and Tremelimumab combination, a 2-step safety run-in phase has been performed before the randomised phase II. The safety run-in phase did not show any safety issue and the randomised phase II starts in September 2020.
Collapse
Affiliation(s)
- Camille Evrard
- Service d'Oncologie Médicale, CHU de Poitiers, Poitiers, France
| | - Christophe Louvet
- Service d'Oncologie Médicale, Institut Mutualiste Montsouris, Paris, France
| | - Farid El Hajbi
- Service de Cancérologie Digestive, Centre Oscar Lambret, Lille, France
| | - Frédéric DI Fiore
- Service d'Hépato-gastroentérologie, Hôpital universitaire de Rouen, Université de Normandie, UNIROUEN, Inserm 1245, IRON group, Rouen 76000, France
| | - Karine LE Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Université de Bourgogne Franche-Comté Dijon, France
| | - Thomas Aparicio
- Service d'Hépato-gastroentérologie, Hôpital Saint Louis, AP-HP, Paris, France
| | - Olivier Bouché
- Service de Cancérologie Digestive, CHU de Reims, Reims, France
| | - Pierre Laurent-Puig
- INSERM U775, Faculté des Sciences Fondamentales et Biomédicales, Centre Universitaire des Saints-Pères, Université des Saints Pères, Paris Descartes, Paris, France
| | - Frédéric Bibeau
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Normandie Université, Caen, France
| | - Thierry Lecomte
- Service d'Hépato-gastroentérologie, CHU de Tours, Tours, France
| | - Astrid Lièvre
- Service des Maladies de l'Appareil Digestif, CHU Pontchaillou, Université de Rennes 1, INSERM U1242, Rennes, France
| | - Rosine Guimbaud
- Service d'Oncologie Médicale, Pôle Digestif, CHU de Toulouse, Toulouse, France
| | - Stefano Kim
- Service d'Oncologie Médicale, CHRU Jean Minjoz, Besançon, France
| | - Aziz Zaanan
- Service de Gastroentérologie et d'Oncologie Digestive, Hôpital Européen George Pompidou, Université de Paris, AP-HP, Paris, France
| | - Harry Sokol
- Université de la Sorbonne, INSERM, Centre de Recherche Paris Saint-Antoine, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Paris, France; Université de Paris-Saclay, INRAE, AgroParisTech, Institut Micalis, Jouy-en-Josas, France; Paris Center for Microbiome Medicine (PaCeMM), Paris, France
| | - Benoist Chibaudel
- Service d'Oncologie Médicale, Hôpital Franco-Britannique - Fondation Cognacq-Jay, Levallois Perret, France
| | - Jérome Desrame
- Service d'Oncologie Médicale, Hôpital privé Jean Mermoz, Lyon, France
| | - Sabrina Pierre
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Université de Bourgogne Franche-Comté Dijon, France
| | - Daniel Gonzalez
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Université de Bourgogne Franche-Comté Dijon, France
| | - Come Lepage
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Université de Bourgogne Franche-Comté Dijon, France; Service d'Hépato-gastroentérologie, CHU de Dijon, France
| | - David Tougeron
- Service d'Oncologie Médicale, CHU de Poitiers, Poitiers, France; Service d'Hépato-gastroentérologie, CHU de Poitiers et Université de Poitiers, 2 rue de la Milétrie, Poitiers 86021, France.
| |
Collapse
|
810
|
Shitara K, Tabernero J. Quantifying the Long-term Survival Benefit of Pembrolizumab for Patients With Advanced Gastric Cancer-Reply. JAMA Oncol 2021; 7:633. [PMID: 33538773 DOI: 10.1001/jamaoncol.2020.8011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Kohei Shitara
- National Cancer Center Hospital East, Kashiwa-Shi, Japan
| | - Josep Tabernero
- Vall d'Hebron University Hospital (HUVH) and Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
811
|
Wainberg ZA, Fuchs CS, Tabernero J, Shitara K, Muro K, Van Cutsem E, Bang YJ, Chung HC, Yamaguchi K, Varga E, Chen JS, Hochhauser D, Thuss-Patience P, Al-Batran SE, Garrido M, Kher U, Shih CS, Shah S, Bhagia P, Chao J. Efficacy of Pembrolizumab Monotherapy for Advanced Gastric/Gastroesophageal Junction Cancer with Programmed Death Ligand 1 Combined Positive Score ≥10. Clin Cancer Res 2021; 27:1923-1931. [PMID: 33446564 DOI: 10.1158/1078-0432.ccr-20-2980] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/16/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Pembrolizumab demonstrated efficacy in PD-L1-positive [combined positive score (CPS) ≥1] advanced gastric/gastroesophageal junction (G/GEJ) cancer in the first-, second-, and third-line setting in KEYNOTE-062, KEYNOTE-061, and KEYNOTE-059, respectively. To better delineate the specificity of CPS as a predictor of clinical outcomes, we analyzed pembrolizumab efficacy in patients with CPS ≥ 10 in these trials. PATIENTS AND METHODS Included were patients with CPS ≥ 10 tumors from KEYNOTE-059 cohort 1 (pembrolizumab, n = 46; post hoc), KEYNOTE-061 (pembrolizumab, n = 53; chemotherapy, n = 55; post hoc), and KEYNOTE-062 (pembrolizumab, n = 92; chemotherapy, n = 90; primary). Efficacy outcomes were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and duration of response (DOR). RESULTS In KEYNOTE-059, median follow-up was 6 months, median OS was 8 months [95% confidence interval (CI), 5.8-11.1], ORR was 17%, and median (range) DOR was 21 months (3+ to 35+). In KEYNOTE-061, median follow-up was 9 months, median OS (pembrolizumab vs. chemotherapy) was 10 versus 8 months (HR, 0.64; 95% CI, 0.41-1.02), median PFS was 3 months versus 3 months (HR, 0.86; 95% CI, 0.56-1.33), ORR was 25% versus 9%, and median (range) DOR was not reached (4 to 26+ months) versus 7 months (3-7). In KEYNOTE-062, median follow-up was 11 months, median OS (pembrolizumab vs. chemotherapy) was 17 months versus 11 months (HR, 0.69; 95% CI, 0.49-0.97), median PFS was 3 months versus 6 months (HR, 1.09, 95% CI; 0.79-1.49), ORR was 25% versus 38%, and median (range) DOR was 19 months (1+ to 34+) versus 7 months (2+ to 30+). CONCLUSIONS This comprehensive analysis showed consistent improvements toward more favorable clinical outcomes with pembrolizumab across lines of therapy in patients with CPS ≥ 10 G/GEJ cancer.
Collapse
Affiliation(s)
- Zev A Wainberg
- Department of the Division of Hematology Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Charles S Fuchs
- Department of Medical Oncology, Yale Cancer Center, Smilow Cancer Hospital, New Haven, Connecticut
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Hyun Cheol Chung
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea (South)
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Eniko Varga
- Department of Oncology, Debreceni Egyetem Klinikai Központ, Debrecen, Hungary
| | - Jen-Shi Chen
- Department of Hematology, Oncology, Linkou Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Daniel Hochhauser
- Department of Medical Oncology, UCL Cancer Institute, London, United Kingdom
| | - Peter Thuss-Patience
- Department of Hematology, Oncology, and Tumor Immunology, Charité-University Medicine Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | | | - Marcelo Garrido
- Department of Hematology and Medical Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Uma Kher
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, New Jersey
| | - Chie-Schin Shih
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, New Jersey
| | - Sukrut Shah
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, New Jersey
| | - Pooja Bhagia
- Department of Medical Oncology, Merck & Co., Inc., Kenilworth, New Jersey
| | - Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
812
|
Ghidini M, Petrillo A, Botticelli A, Trapani D, Parisi A, La Salvia A, Sajjadi E, Piciotti R, Fusco N, Khakoo S. How to Best Exploit Immunotherapeutics in Advanced Gastric Cancer: Between Biomarkers and Novel Cell-Based Approaches. J Clin Med 2021; 10:1412. [PMID: 33915839 PMCID: PMC8037391 DOI: 10.3390/jcm10071412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Despite extensive research efforts, advanced gastric cancer still has a dismal prognosis with conventional treatment options. Immune checkpoint inhibitors have revolutionized the treatment landscape for many solid tumors. Amongst gastric cancer subtypes, tumors with microsatellite instability and Epstein Barr Virus positive tumors provide the strongest rationale for responding to immunotherapy. Various predictive biomarkers such as mismatch repair status, programmed death ligand 1 expression, tumor mutational burden, assessment of tumor infiltrating lymphocytes and circulating biomarkers have been evaluated. However, results have been inconsistent due to different methodologies and thresholds used. Clinical implementation therefore remains a challenge. The role of immune checkpoint inhibitors in gastric cancer is emerging with data from monotherapy in the heavily pre-treated population already available and studies in earlier disease settings with different combinatorial approaches in progress. Immune checkpoint inhibitor combinations with chemotherapy (CT), anti-angiogenics, tyrosine kinase inhibitors, anti-Her2 directed therapy, poly (ADP-ribose) polymerase inhibitors or dual checkpoint inhibitor strategies are being explored. Moreover, novel strategies including vaccines and CAR T cell therapy are also being trialed. Here we provide an update on predictive biomarkers for response to immunotherapy with an overview of their strengths and limitations. We discuss clinical trials that have been reported and trials in progress whilst providing an account of future steps needed to improve outcome in this lethal disease.
Collapse
Affiliation(s)
- Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University, 00189 Rome, Italy;
- Medical Oncology (B), Policlinico Umberto I, 00161 Rome, Italy
| | - Dario Trapani
- Division of Early Drug Development for innovative therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy;
| | - Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy
| | - Anna La Salvia
- Department of Oncology, University Hospital 12 De Octubre, 28041 Madrid, Spain;
| | - Elham Sajjadi
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Roberto Piciotti
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Shelize Khakoo
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM25PT, UK;
| |
Collapse
|
813
|
McCaw ZR, Ludmir EB, Wei LJ. Quantifying the Long-term Survival Benefit of Pembrolizumab for Patients With Advanced Gastric Cancer. JAMA Oncol 2021; 7:632-633. [PMID: 33538772 DOI: 10.1001/jamaoncol.2020.8002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
| | - Ethan B Ludmir
- The University of Texas MD Anderson Cancer Center, Houston
| | - Lee-Jen Wei
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
814
|
Mamdani H, Jalal SI. Where to Start and What to Do Next: The Sequencing of Treatments in Metastatic Esophagogastric Cancer. Am Soc Clin Oncol Educ Book 2021; 41:1-16. [PMID: 33770461 DOI: 10.1200/edbk_321243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Esophagogastric cancer is associated with rising incidence and high mortality. Nearly 40% of patients have metastatic disease at the time of diagnosis with poor 5-year overall survival. The treatment of squamous cell carcinoma of the esophagus and gastroesophageal adenocarcinoma has started to bifurcate in recent years, owing to the evolving understanding of the biologic and genomic characteristics of these tumors. Incorporation of HER2-directed therapy in the form of monoclonal antibody and antibody-drug conjugate is now standard of care for patients with HER2-positive disease. The addition of immune checkpoint inhibitors to the therapeutic landscape of metastatic esophagogastric cancer is associated with modest improvement in overall survival, and definition of predictive biomarkers of response to checkpoint inhibition remains imprecise. A number of therapeutic targets including FGFR2b, Claudin 18.2, DKK-1, and DNA repair defects are being explored in clinical trials. Similarly, combination immunotherapy and novel HER2-targeting agents, such as bispecific antibody and small-molecule inhibitors, are at various stages of clinical development. Despite the progress made in the field of targeted therapies and checkpoint inhibition, chemotherapy remains an integral part of treatment of metastatic esophagogastric cancer but is associated with considerable toxicity. Clinical trials focusing on minimizing toxicity of currently available therapeutic agents, development of novel biomarker-driven treatment strategies, and overcoming resistance to immune checkpoint inhibition will define the future of this traditionally indelible disease.
Collapse
Affiliation(s)
- Hirva Mamdani
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Shadia I Jalal
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| |
Collapse
|
815
|
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: 176] [Impact Index Per Article: 44.0] [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
|
816
|
Fan X, Wang D, Zhang W, Liu J, Liu C, Li Q, Ma Z, Li H, Guan X, Bai Y, Yang J, Lou C, Li X, Wang G, Li Z. Inflammatory Markers Predict Survival in Patients With Advanced Gastric and Colorectal Cancers Receiving Anti-PD-1 Therapy. Front Cell Dev Biol 2021; 9:638312. [PMID: 33791296 PMCID: PMC8005614 DOI: 10.3389/fcell.2021.638312] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
There is a lack of useful biomarkers for predicting the efficacy of anti-programmed death-1 (PD-1) therapy for advanced gastric and colorectal cancer. To address this issue, in this study we investigated the correlation between inflammatory marker expression and survival in patients with advanced gastric and colorectal cancer. Data for 111 patients with advanced gastric and colorectal cancer treated with anti-PD-1 regimens were retrospectively analyzed. Neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), and clinical characteristics of each patient were selected as the main variables. Overall response rate, disease control rate, and progression-free survival were primary endpoints, and overall survival and immune-related adverse events (irAEs) were secondary endpoints. The chi-squared test and Fisher's exact test were used to evaluate relationships between categorical variables. Uni- and multivariate Cox regression analyses were performed, and median progression-free survival and overall survival were estimated with the Kaplan-Meier method. The overall response rate and disease control rate of anti-PD-1therapy in advanced gastric and colorectal tumors were 12.61 and 66.66%, respectively. The patients with MLR < 0.31, NLR < 5, and PLR < 135 had a significantly higher disease control rate than those with MLR > 0.31, NLR > 5, and PLR > 135 (P < 0.05). The multivariate analysis revealed that MLR < 0.31, BMI > 18.5, and anti-PD-1 therapy in first-line were associated with prolonged PFS. MLR < 0.31 and BMI > 18.5 were associated with prolonged overall survival. The irAE rate differed significantly between PLR groups, and PLR < 135 was associated with an increased rate of irAEs (P = 0.028). These results indicate that the inflammatory markers NLR, MLR, and PLR have clinical utility for predicting survival or risk of irAEs in patients with advanced gastric cancer and colorectal cancer.
Collapse
Affiliation(s)
- Xiaona Fan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dan Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenjing Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jinshuang Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qingwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhigang Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hengzhen Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Guan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yibing Bai
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiani Yang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Changjie Lou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaobo Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhiwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
817
|
Salas-Benito D, Pérez-Gracia JL, Ponz-Sarvisé M, Rodriguez-Ruiz ME, Martínez-Forero I, Castañón E, López-Picazo JM, Sanmamed MF, Melero I. Paradigms on Immunotherapy Combinations with Chemotherapy. Cancer Discov 2021; 11:1353-1367. [PMID: 33712487 DOI: 10.1158/2159-8290.cd-20-1312] [Citation(s) in RCA: 247] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/21/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022]
Abstract
Checkpoint inhibitors are being added to standard-of-care chemotherapy in multiple clinical trials. Success has been reported in non-small and small cell lung carcinomas and urothelial, head and neck, gastric, and esophageal cancers, and promising results are already available in triple-negative breast and pancreatic malignancies. The potential mechanisms of synergy include immunogenic tumor cell death, antiangiogenesis, selective depletion of myeloid immunosuppressive cells, and lymphopenia, which reduces regulatory T cells and makes room for proliferation of effector T cells. However, chemotherapy regimens have not been optimized for such combinations, perhaps explaining some recent clinical trial disappointments. Approaches to make the most of chemoimmunotherapy include neoadjuvant and adjuvant schemes.Significance: Immunotherapy of cancer based on PD-1/PD-L1 blockade has prompted a revolution in cancer clinical management. Evidence in phase III clinical trials already supports combinations of immunotherapy with standard-of-care chemotherapy for a number of malignant diseases. This review focuses on such evidence and provides an overview of the potential synergistic mechanisms of action and the opportunities to optimize chemoimmunotherapy regimens.
Collapse
Affiliation(s)
- Diego Salas-Benito
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain. .,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - José L Pérez-Gracia
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Ponz-Sarvisé
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - María E Rodriguez-Ruiz
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Eduardo Castañón
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - José M López-Picazo
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Miguel F Sanmamed
- Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain.,Center for Medical Applied Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Biomedical Research Network in Oncology (CIBERONC), Pamplona, Spain
| | - Ignacio Melero
- Clinical Trials Unit, Clínica Universidad de Navarra, Pamplona, Spain. .,Center for Medical Applied Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Biomedical Research Network in Oncology (CIBERONC), Pamplona, Spain.,Immunology Department, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
818
|
Rocha-Filho DR, Peixoto RD, Weschenfelder RF, Rego JFM, Riechelmann R, Coutinho AK, Fernandes GS, Jacome AA, Andrade AC, Murad AM, Mello CAL, Miguel DSCG, Gomes DBD, Racy DJ, Moraes ED, Akaishi EH, Carvalho ES, Mello ES, Filho FM, Coimbra FJF, Capareli FC, Arruda FF, Vieira FMAC, Takeda FR, Cotti GCC, Pereira GLS, Paulo GA, Ribeiro HSC, Lourenco LG, Crosara M, Toneto MG, Oliveira MB, de Lourdes Oliveira M, Begnami MD, Forones NM, Yagi O, Ashton-Prolla P, Aguillar PB, Amaral PCG, Hoff PM, Araujo RLC, Di Paula Filho RP, Gansl RC, Gil RA, Pfiffer TEF, Souza T, Ribeiro U, Jesus VHF, Costa WL, Prolla G. Brazilian Group of Gastrointestinal Tumours' consensus guidelines for the management of oesophageal cancer. Ecancermedicalscience 2021; 15:1195. [PMID: 33889204 PMCID: PMC8043684 DOI: 10.3332/ecancer.2021.1195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Indexed: 11/28/2022] Open
Abstract
Oesophageal cancer is among the ten most common types of cancer worldwide. More than 80% of the cases and deaths related to the disease occur in developing countries. Local socio-economic, epidemiologic and healthcare particularities led us to create a Brazilian guideline for the management of oesophageal and oesophagogastric junction (OGJ) carcinomas. The Brazilian Group of Gastrointestinal Tumours invited 50 physicians with different backgrounds, including radiology, pathology, endoscopy, nuclear medicine, genetics, oncological surgery, radiotherapy and clinical oncology, to collaborate. This document was prepared based on an extensive review of topics related to heredity, diagnosis, staging, pathology, endoscopy, surgery, radiation, systemic therapy (including checkpoint inhibitors) and follow-up, which was followed by presentation, discussion and voting by the panel members. It provides updated evidence-based recommendations to guide clinical management of oesophageal and OGJ carcinomas in several scenarios and clinical settings.
Collapse
Affiliation(s)
- Duilio R Rocha-Filho
- Hospital Universitário Walter Cantídio, 60430-372 Fortaleza, Brazil
- Grupo Oncologia D’Or, 04535-110 São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | - Diogo B D Gomes
- Hospital Israelita Albert Einstein, 05652-900, São Paulo, Brazil
| | - Douglas J Racy
- Hospital Beneficência Portuguesa de São Paulo, 01323-001 São Paulo, Brazil
| | | | - Eduardo H Akaishi
- Faculdade de Medicina da Universidade de São Paulo, 01246903 São Paulo, Brazil
| | | | - Evandro S Mello
- Faculdade de Medicina da Universidade de São Paulo, 01246903 São Paulo, Brazil
| | - Fauze Maluf Filho
- Faculdade de Medicina da Universidade de São Paulo, 01246903 São Paulo, Brazil
| | | | | | | | | | - Flavio R Takeda
- Faculdade de Medicina da Universidade de São Paulo, 01246903 São Paulo, Brazil
| | | | | | - Gustavo A Paulo
- Universidade Federal de São Paulo, 04040-003 São Paulo, Brazil
| | | | | | | | | | - Marcos B Oliveira
- Faculdade de Ciências Médicas da Santa Casa de São Paulo, 01238-010 São Paulo, Brazil
| | | | | | - Nora M Forones
- Universidade Federal de São Paulo, 04040-003 São Paulo, Brazil
| | - Osmar Yagi
- Faculdade de Medicina da Universidade de São Paulo, 01246903 São Paulo, Brazil
| | | | | | | | - Paulo M Hoff
- Grupo Oncologia D’Or, 04535-110 São Paulo, Brazil
| | | | | | | | | | | | - Tulio Souza
- Hospital Aliança de Salvador, 41920-900 Salvador, Brazil
| | - Ulysses Ribeiro
- Faculdade de Medicina da Universidade de São Paulo, 01246903 São Paulo, Brazil
| | | | | | | |
Collapse
|
819
|
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
|
820
|
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
|
821
|
Smyth EC, Gambardella V, Cervantes A, Fleitas T. Checkpoint inhibitors for gastroesophageal cancers: dissecting heterogeneity to better understand their role in first-line and adjuvant therapy. Ann Oncol 2021; 32:590-599. [PMID: 33609722 DOI: 10.1016/j.annonc.2021.02.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
Gastroesophageal adenocarcinoma (GEA) and squamous esophageal cancer (ESCC) are responsible for >1 million deaths annually globally. Until now, patients with metastatic GEA and ESCC could anticipate survival of <1 year. Anti- programmed cell death protein 1 (anti-PD-1) monotherapy has demonstrated modest efficacy in previously treated GEA and ESCC. In 2020, four pivotal trials have established anti-PD-1 therapy as a new standard of care for selected GEA and ESCC patients as first-line advanced and adjuvant therapy. In this review, we discuss the recent results of the CheckMate 649, ATTRACTION-4, KEYNOTE-590 and CheckMate 577 trials. We consider these results in the context of current standards of care and historical trials of immune checkpoint blockade in GEA and ESCC. We explore biomarker selection for anti-PD-1 therapy and appraise the future of combination therapies. In CheckMate 649, treatment with oxaliplatin-fluoropyrimidine chemotherapy plus nivolumab in patients with combined positive score ≥5 GEA tumors provided a clinically meaningful and statistically significant improvement in overall survival. The ATTRACTION-4 trial did not see a similar overall survival benefit, despite a clear improvement in progression-free survival. We review potential explanations for this result. KEYNOTE-590 showed profoundly improved survival when pembrolizumab was added to cisplatin-fluoropyrimidine chemotherapy in ESCC patients with combined positive score ≥10 tumors; this benefit was less convincing in unselected ESCC. Finally, CheckMate 577 provides proof-of-concept for the improvement in disease-free survival with adjuvant nivolumab in high-risk resected GEA and ESCC following trimodality therapy. Immune checkpoint blockade has come of age in GEA and ESCC, and will now be integrated into first-line and earlier lines of therapy, providing benefit for a larger proportion of patients. Biomarker standardization will be critical to select the patients most likely to benefit from treatment. For patients with immune evasive tumors, novel combinations under development show promise; however, global trials are needed.
Collapse
Affiliation(s)
- E C Smyth
- Cambridge University Hospitals National Health Service Foundation Trust, Department of Oncology, Cambridge, UK
| | - V Gambardella
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - A Cervantes
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| | - T Fleitas
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
822
|
Nakajima TE, Kadowaki S, Minashi K, Nishina T, Yamanaka T, Hayashi Y, Izawa N, Muro K, Hironaka S, Kajiwara T, Kawakami Y. Multicenter Phase I/II Study of Nivolumab Combined with Paclitaxel Plus Ramucirumab as Second-line Treatment in Patients with Advanced Gastric Cancer. Clin Cancer Res 2021; 27:1029-1036. [PMID: 33262133 DOI: 10.1158/1078-0432.ccr-20-3559] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We conducted a phase I/II study to investigate the safety and efficacy of nivolumab with paclitaxel plus ramucirumab. PATIENTS AND METHODS Patients with advanced gastric cancer (AGC) refractory to first-line chemotherapy were included. Patients received nivolumab (3 mg/kg on days 1 and 15) combined with paclitaxel (80 mg/m2 on days 1, 8, and 15) and ramucirumab (8 mg/kg on days 1 and 15) every 4 weeks. After feasibility evaluation in six patients (phase I), 37 additional patients were enrolled in the phase II part with the primary endpoint of 6-month progression-free survival (PFS) rate with two-sided 80% confidence interval (CI). The combined positive score (CPS) was defined as the number of programmed death-ligand 1-positive cells divided by the total number of viable tumor cells multiplied by 100. RESULTS Forty-three patients were enrolled. Of these, 60.5% had CPS ≥ 1. Dose-limiting toxicities were observed in two patients, and the recommended dose was determined as level 1. Thirty-nine (90.7%) patients experienced treatment-related adverse events (AEs) grade ≥3 and 14 (32.6%) patients experienced immune-related AEs grade ≥3. The overall response rate was 37.2% (95% CI, 23.0%-53.5%) and the 6-month PFS rate was 46.5% (80% CI, 36.4%-55.8%; P = 0.067). Median survival time was 13.1 months (95% CI, 8.0-16.6 months): 13.8 months (95% CI, 8.0-19.5 months) in patients with CPS ≥ 1 and 8.0 months (95% CI, 4.8-24.1 months) in patients with CPS < 1. CONCLUSIONS Nivolumab with paclitaxel plus ramucirumab demonstrated promising antitumor activity with manageable toxicities as second-line treatment for AGC.
Collapse
Affiliation(s)
- Takako Eguchi Nakajima
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan.
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy, Kyoto University Hospital, Kyoto City, Kyoto, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Keiko Minashi
- Department of Clinical Trial Promotion, Chiba Cancer Center, Chuo, Chiba, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Yuichiro Hayashi
- Division of Pathology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Naoki Izawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Shuichi Hironaka
- Department of Clinical Trial Promotion, Chiba Cancer Center, Chuo, Chiba, Japan
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- Department of Immunology, International University of Health and Welfare School of Medicine, Narita, Chiba, Japan
| |
Collapse
|
823
|
Kawakami H, Hironaka S, Esaki T, Chayama K, Tsuda M, Sugimoto N, Kadowaki S, Makiyama A, Machida N, Hirano H, Hirata K, Hara H, Yabusaki H, Komatsu Y, Muro K. An Investigator-Initiated Phase 2 Study of Nivolumab Plus Low-Dose Ipilimumab as First-Line Therapy for Microsatellite Instability-High Advanced Gastric or Esophagogastric Junction Cancer (NO LIMIT, WJOG13320G/CA209-7W7). Cancers (Basel) 2021; 13:805. [PMID: 33671871 PMCID: PMC7918984 DOI: 10.3390/cancers13040805] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 12/30/2022] Open
Abstract
Nivolumab (NIVO) plus low-dose ipilimumab (IPI) has shown a promising survival benefit in first-line treatment of microsatellite instability-high (MSI-H) colorectal cancer. We hypothesized that this regimen might also be beneficial for MSI-H gastric cancer (GC), which accounts for ~5% of all GC cases. NO LIMIT (WJOG13320G/CA209-7W7) is an investigator-initiated, single-arm, open-label, 14-center phase 2 trial of NIVO plus low-dose IPI for MSI-H GC in the first-line setting. Eligibility criteria include unresectable advanced, recurrent, or metastatic gastric or esophagogastric junction cancer with a histologically confirmed diagnosis of adenocarcinoma; confirmed MSI-H status with the MSI-IVD Kit (FALCO); no prior systemic anticancer therapy; an Eastern Cooperative Oncology Group performance status of 0 or 1; and a measurable lesion per RECIST 1.1. The primary objective of the study is to determine the overall response rate (ORR) for the NIVO+IPI regimen as assessed by blinded independent central review. Secondary end points include progression-free survival, overall survival, duration of response, safety, tolerability, and biomarkers. The number of patients was set at 28 on the basis of the threshold and expected ORR values of 35 and 65%, respectively, with a one-sided alpha error of 0.025 and power of 0.80. Subjects will receive treatment with nivolumab (240 mg) biweekly in combination with ipilimumab (1 mg/kg) every 6 weeks. The results of this study should clarify the therapeutic potential of NIVO+IPI for MSI-H GC in the first-line setting. Trial registration: JapicCTI-205400.
Collapse
Affiliation(s)
- Hisato Kawakami
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
| | - Shuichi Hironaka
- Department of Medical Oncology and Hematology, Faculty of Medicine, Oita University, Oita 879-5593, Japan;
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan;
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Masahiro Tsuda
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Hyogo 673-8558, Japan;
| | - Naotoshi Sugimoto
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka 541-8567, Japan;
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Aichi 464-8681, Japan; (S.K.); (K.M.)
| | | | - Nozomu Machida
- Department of Gastroenterology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Hidekazu Hirano
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan;
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama 362-0806, Japan;
| | - Hiroshi Yabusaki
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, Niigata 951-8566, Japan;
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Hokkaido 060-8648, Japan;
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Aichi 464-8681, Japan; (S.K.); (K.M.)
| |
Collapse
|
824
|
Rodriguez A, Esposito F, Oliveres H, Torres F, Maurel J. Are Quality of Randomized Clinical Trials and ESMO-Magnitude of Clinical Benefit Scale Two Sides of the Same Coin, to Grade Recommendations for Drug Approval? J Clin Med 2021; 10:746. [PMID: 33668473 PMCID: PMC7918206 DOI: 10.3390/jcm10040746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/06/2021] [Accepted: 02/11/2021] [Indexed: 12/20/2022] Open
Abstract
The approval of a new drug for cancer treatment by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) is based on positive, well-designed randomized phase III clinical trials (RCTs). However, not all of them are analyzed to support the recommendations. For this reason, there are different scales to quantify and evaluate the quality of RCTs and the magnitude of the clinical benefits of new drugs for treating solid tumors. In this review, we discuss the value of the progression-free survival (PFS) as an endpoint in RCTs and the concordance between it and the overall survival (OS) as a measure of the quality of clinical trial designs. We summarize and analyze the different scales to evaluate the clinical benefits of new drugs such as the The American Society of Clinical Oncology value framework (ASCO-VF-NHB16) and European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS) and the concordance between them, focusing on metastatic colorectal cancer (mCRC). We propose several definitions that would help to evaluate the quality of RCT, the magnitude of clinical benefit and the appropriate approval of new drugs in oncology.
Collapse
Affiliation(s)
- Adela Rodriguez
- Department of Medical Oncology, Hospital Clinic of Barcelona,08036 Barcelona, Spain; (A.R.); (F.E.); (H.O.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Medical Statistics Core Facility, IDIBAPS, Hospital Clinic, 08036 Barcelona, Spain
| | - Francis Esposito
- Department of Medical Oncology, Hospital Clinic of Barcelona,08036 Barcelona, Spain; (A.R.); (F.E.); (H.O.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Medical Statistics Core Facility, IDIBAPS, Hospital Clinic, 08036 Barcelona, Spain
| | - Helena Oliveres
- Department of Medical Oncology, Hospital Clinic of Barcelona,08036 Barcelona, Spain; (A.R.); (F.E.); (H.O.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Medical Statistics Core Facility, IDIBAPS, Hospital Clinic, 08036 Barcelona, Spain
| | - Ferran Torres
- Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, 08036 Barcelona, Spain
| | - Joan Maurel
- Department of Medical Oncology, Hospital Clinic of Barcelona,08036 Barcelona, Spain; (A.R.); (F.E.); (H.O.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Medical Statistics Core Facility, IDIBAPS, Hospital Clinic, 08036 Barcelona, Spain
| |
Collapse
|
825
|
Högner A, Thuss-Patience P. Immune Checkpoint Inhibition in Oesophago-Gastric Carcinoma. Pharmaceuticals (Basel) 2021; 14:151. [PMID: 33673374 PMCID: PMC7918118 DOI: 10.3390/ph14020151] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors enrich the therapeutic landscape in oesophago-gastric carcinoma. With regard to oesophageal squamous cell carcinoma (ESCC), the selective PD-1 (programmed cell death receptor 1)-inhibitor nivolumab improves disease-free survival in the adjuvant therapy setting (CHECKMATE-577). In first-line treatment, ESCC patients (pts) benefit in overall survival (OS) from the PD-1-inhibitor pembrolizumab in combination with chemotherapy (KEYNOTE-590). In the second-line setting, nivolumab (ATTRACTION-03) and pembrolizumab (KEYNOTE-181) demonstrate a benefit in OS compared with chemotherapy. These data resulted in the approval of nivolumab for the second-line treatment of advanced ESCC pts regardless of PD-L1 (programmed cell death ligand 1) status in Europe, Asia, and the USA, and pembrolizumab for pts with PD-L1 CPS (combined positivity score) ≥ 10 in Asia and the USA. Further approvals can be expected. In gastro-oesophageal junction and gastric cancer, the addition of nivolumab to chemotherapy in first-line treatment improves OS in pts with advanced disease with PD-L1 CPS ≥ 5 (CHECKMATE-649). Additionally, pembrolizumab was non-inferior to chemotherapy for OS in PD-L1 CPS ≥ 1 pts (KEYNOTE-062). In third-line treatment, nivolumab shows benefits in OS regardless of PD-L1 expression (ATTRACTION-02) with approval in Asia, and pembrolizumab prolonged the duration of response in PD-L1 positive pts (KEYNOTE-059) with approval in the USA. We discuss the recent results of the completed phase II and III clinical trials.
Collapse
Affiliation(s)
| | - Peter Thuss-Patience
- Campus Virchow-Klinikum, Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany;
| |
Collapse
|
826
|
Zhu M, Jin Z, Hubbard JM. Management of Non-Colorectal Digestive Cancers with Microsatellite Instability. Cancers (Basel) 2021; 13:651. [PMID: 33561950 PMCID: PMC7915546 DOI: 10.3390/cancers13040651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/26/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
Microsatellite instability (MSI) is a hallmark of genetic predisposition to DNA damage. It arises from either germline or somatic events leading to impaired function of the mismatch repair system. It can be detected via genetic sequencing or immunohistochemistry with relatively high concordance rates. The presence of MSI in a tumor reflects a high neoantigen load and predicts favorable treatment response to immune checkpoint inhibitors (ICIs). In gastrointestinal cancers, MSI is a predictive biomarker for ICIs with potential prognostic impact but its clinical utility varies widely depending on tumor type. This may be explained by the complexity of tumor microenvironment as highlighted by recent translational studies. In this review, we will discuss the predictive and prognostic value of MSI status in non-colorectal cancers of the digestive system, important clinical trials involving ICIs and potential strategies to overcome resistance to immunotherapy.
Collapse
Affiliation(s)
- Mojun Zhu
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA; (Z.J.); (J.M.H.)
| | | | | |
Collapse
|
827
|
Jachetti E, Sangaletti S, Chiodoni C, Ferrara R, Colombo MP. Modulation of PD-1/PD-L1 axis in myeloid-derived suppressor cells by anti-cancer treatments. Cell Immunol 2021; 362:104301. [PMID: 33588246 DOI: 10.1016/j.cellimm.2021.104301] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/19/2022]
Abstract
Immuno checkpoint blockade (ICB) targeting the PD-1/PD-L1 axis is the main breakthrough for the treatment of several cancers. Nevertheless, not all patients benefit from this treatment and clinical response not always correlates with PD-L1 expression by tumor cells. The tumor microenvironment, including myeloid derived suppressor cells (MDSCs), can influence therapeutic resistance to ICB. MDSCs also express PD-L1, which contributes to their suppressive activity. Moreover, anticancer therapies including chemotherapy, radiotherapy, hormone- and targeted- therapies can modulate MDSCs recruitment, activity and PD-L1 expression. Such effects can be induced also by innovative anticancer treatments targeting metabolism and lifestyle. The outcome on cancer progression can be either positive or negative, depending on tumor type, treatment schedule and possible combination with ICB. Further studies are needed to better understand the effects of cancer therapies on the PD-1/PD-L1 axis, to identify patients that could benefit from combinatorial regimens including ICB or that rather should avoid it.
Collapse
Affiliation(s)
- Elena Jachetti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sabina Sangaletti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Chiodoni
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberto Ferrara
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
828
|
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: 129] [Impact Index Per Article: 32.3] [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
|
829
|
Kim W, Chu TH, Nienhüser H, Jiang Z, Portillo AD, Remotti HE, White RA, Hayakawa Y, Tomita H, Fox JG, Drake CG, Wang TC. PD-1 Signaling Promotes Tumor-Infiltrating Myeloid-Derived Suppressor Cells and Gastric Tumorigenesis in Mice. Gastroenterology 2021; 160:781-796. [PMID: 33129844 PMCID: PMC7878361 DOI: 10.1053/j.gastro.2020.10.036] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Immune checkpoint inhibitors have limited efficacy in many tumors. We investigated mechanisms of tumor resistance to inhibitors of programmed cell death-1 (PDCD1, also called PD-1) in mice with gastric cancer, and the role of its ligand, PD-L1. METHODS Gastrin-deficient mice were given N-methyl-N-nitrosourea (MNU) in drinking water along with Helicobacter felis to induce gastric tumor formation; we also performed studies with H/K-ATPase-hIL1B mice, which develop spontaneous gastric tumors at the antral-corpus junction and have parietal cells that constitutively secrete interleukin 1B. Mice were given injections of an antibody against PD-1 or an isotype control before tumors developed, or anti-PD-1 and 5-fluorouracil and oxaliplatin, or an antibody against lymphocyte antigen 6 complex locus G (also called Gr-1), which depletes myeloid-derived suppressor cells [MDSCs]), after tumors developed. We generated knock-in mice that express PD-L1 specifically in the gastric epithelium or myeloid lineage. RESULTS When given to gastrin-deficient mice before tumors grew, anti-PD-1 significantly reduced tumor size and increased tumor infiltration by T cells. However, anti-PD-1 alone did not have significant effects on established tumors in these mice. Neither early nor late anti-PD-1 administration reduced tumor growth in the presence of MDSCs in H/K-ATPase-hIL-1β mice. The combination of 5-fluorouracil and oxaliplatin reduced MDSCs, increased numbers of intra-tumor CD8+ T cells, and increased the response of tumors to anti-PD-1; however, this resulted in increased tumor expression of PD-L1. Expression of PD-L1 by tumor or immune cells increased gastric tumorigenesis in mice given MNU. Mice with gastric epithelial cells that expressed PD-L1 did not develop spontaneous tumors, but they developed more and larger tumors after administration of MNU and H felis, with accumulation of MDSCs. CONCLUSIONS In mouse models of gastric cancer, 5-fluorouracil and oxaliplatin reduce numbers of MDSCs to increase the effects of anti-PD-1, which promotes tumor infiltration by CD8+ T cells. However, these chemotherapeutic agents also induce expression of PD-L1 by tumor cells. Expression of PD-L1 by gastric epithelial cells increases tumorigenesis in response to MNU and H felis, and accumulation of MDSCs, which promote tumor progression. The timing and site of PD-L1 expression is therefore important in gastric tumorigenesis and should be considered in design of therapeutic regimens.
Collapse
Affiliation(s)
- Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Timothy H. Chu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Armando Del Portillo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Helen E. Remotti
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Ruth A. White
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 01239, USA
| | - Charles G. Drake
- Division of Hematology and Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
830
|
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: 167] [Impact Index Per Article: 41.8] [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
|
831
|
Pietrantonio F, Randon G, Di Bartolomeo M, Luciani A, Chao J, Smyth EC, Petrelli F. Predictive role of microsatellite instability for PD-1 blockade in patients with advanced gastric cancer: a meta-analysis of randomized clinical trials. ESMO Open 2021; 6:100036. [PMID: 33460964 PMCID: PMC7815473 DOI: 10.1016/j.esmoop.2020.100036] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Several post hoc analyses of randomized controlled trials (RCTs) suggested the importance of microsatellite instability (MSI) as a positive predictive factor to immunotherapy in patients with advanced gastric cancer (GC); however, individually these have low statistical power. METHODS RCTs investigating treatment with or without an anti-programmed cell death protein 1 (PD-1) agent for advanced GC and providing outcome according to MSI status were selected. The hazard ratio (HR) and the odds ratio were used to compare the treatment effect on survival outcomes and tumor response, respectively, for anti-PD-1-based therapy compared with standard therapy. Evidence for treatment effect by MSI status was evaluated by a test of interaction. RESULTS The phase III KEYNOTE-062, CheckMate-649, JAVELIN Gastric 100 and KEYNOTE-061 trials were included. A total of 2545 patients with evaluable MSI status were included and 123 (4.8%) had MSI-high cancers. The HR for overall survival benefit with anti-PD-1-based regimens was 0.34 (95% CI: 0.21-0.54) for MSI-high cancers versus 0.85 [95% confidence interval (CI): 0.71-1.00] for microsatellite stable. The treatment effect was significantly different in the two subgroups (P for interaction 0.003). In the MSI-high subgroup, the HR for progression-free survival was 0.57 (95% CI: 0.33-0.97; P = 0.04) and the odds ratio for response was 1.76 (95% CI: 1.10-2.83; P = 0.02). CONCLUSIONS Patients with MSI-high GC should be regarded as a specific and highly immunosensitive population worthy of dedicated clinical trials.
Collapse
Affiliation(s)
- F Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - G Randon
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - A Luciani
- Medical Oncology Unit, Azienda Socio-Sanitaria Territoriale Bergamo Ovest, Treviglio, Bergamo, Italy
| | - J Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, USA
| | - E C Smyth
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - F Petrelli
- Medical Oncology Unit, Azienda Socio-Sanitaria Territoriale Bergamo Ovest, Treviglio, Bergamo, Italy
| |
Collapse
|
832
|
Saeed A, Park R, Sun W. The integration of immune checkpoint inhibitors with VEGF targeted agents in advanced gastric and gastroesophageal adenocarcinoma: a review on the rationale and results of early phase trials. J Hematol Oncol 2021; 14:13. [PMID: 33436042 PMCID: PMC7802258 DOI: 10.1186/s13045-021-01034-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Several targeted therapies have shown efficacy in patients with advanced gastric cancer (GC) and gastroesophageal junction adenocarcinoma (GEJC), including anti-angiogenic agents and immune checkpoint inhibitors. Ramucirumab, an anti-VEGFR2 antibody, has shown efficacy in GC, but the benefits are limited, in part due to MET-mediated resistance. Other VEGF targeted agents like VEGF tyrosine kinase inhibitors (TKIs) with broad multi-kinase inhibitory spectrum like regorafenib and cabozantinib have also shown modest single agent activity in early phase trials. For immune checkpoint inhibitors, pembrolizumab (anti-PD-1) monotherapy confers survival advantage as 3rd line therapy for the PD-L1 expressing GC and GEJC population and has been approved for use in this setting. Extensive tumor microenvironment immune modulatory effects from antiangiogenic agents have been demonstrated from preclinical data which support the clinical study rationale of dual blockade of VEGF and immune checkpoint. In addition, FDA has approved combinations of anti-VEGF/VEGFR with anti-PD-1/PD-L1 agents in hepatocellular carcinoma and renal cell carcinoma. Promising clinical activity has been demonstrated in patients with refractory GC/GEJC when treated with dual blockade combination with antiangiogenic agents and immune checkpoint inhibitors like PD-1/PD-L1 inhibitors in several phase I/II trials. This review highlights the trials investigating these novel combinations as well as their preclinical rationale.
Collapse
Affiliation(s)
- Anwaar Saeed
- Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, 2330 Shawnee Mission Pkwy, Suite 210, Westwood, Kansas City, KS, 66205, USA.
| | - Robin Park
- MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, 2330 Shawnee Mission Pkwy, Suite 210, Westwood, Kansas City, KS, 66205, USA
| |
Collapse
|
833
|
Kawazoe A, Shitara K, Boku N, Yoshikawa T, Terashima M. Current status of immunotherapy for advanced gastric cancer. Jpn J Clin Oncol 2021; 51:20-27. [PMID: 33241322 DOI: 10.1093/jjco/hyaa202] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 09/03/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Recently, immune checkpoint inhibitors such as anti-programmed cell death-1 (PD-1) or programmed cell death ligand-1 (PD-L1) monoclonal antibodies have improved the overall survival of various types of cancers including advanced gastric cancer (AGC). Until now, two ant-PD-1 inhibitors were approved for AGC in Japan: nivolumab as third- or later-line treatment for AGC and pembrolizumab for previously treated patients with microsatellite instability-high tumours. However, a limited number of patients achieved clinical benefit, highlighting the importance of the better selection of patients or additional treatment to overcome resistance to PD-1/PD-L1 blockade. This review focused on pivotal clinical trials, biomarkers and novel combination therapy of immune checkpoint inhibitors forAGC.
Collapse
Affiliation(s)
- Akihito Kawazoe
- Department of Gastrointestinal Medical oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Kohei Shitara
- Department of Gastrointestinal Medical oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo
| | - Takaki Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo
| | | |
Collapse
|
834
|
Kotani D, Shitara K. Trastuzumab deruxtecan for the treatment of patients with HER2-positive gastric cancer. Ther Adv Med Oncol 2021; 13:1758835920986518. [PMID: 33473250 PMCID: PMC7797586 DOI: 10.1177/1758835920986518] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/17/2020] [Indexed: 12/26/2022] Open
Abstract
Trastuzumab deruxtecan (T-DXd) is a novel anti-human epidermal growth
factor receptor 2 (HER2) antibody–drug conjugate composed of a
monoclonal anti-HER2 antibody and a topoisomerase I inhibitor, DX-8951
derivative (an exatecan derivative). T-DXd showed potential anti-tumor
activities in HER2-positive gastric cancer cell lines and xenograft
models. In the randomized, phase II trial DESTINY-Gastric01, T-DXd
demonstrated a significantly higher objective response rate as a
primary endpoint and a longer overall survival as a secondary endpoint
in patients with pretreated HER2-positive advanced gastric cancer
(AGC). Although adverse events caused by T-DXd were generally
manageable, approximately 10% of patients experienced
treatment-related interstitial lung disease. Based on the results of
the DESTINY-Gastric01 trial, T-DXd was approved for HER2-positive
pretreated AGC in Japan. This study reviews the preclinical and
clinical data of T-DXd for treating HER2-positive gastric cancer.
Collapse
Affiliation(s)
- Daisuke Kotani
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| |
Collapse
|
835
|
Boku N, Satoh T, Ryu MH, Chao Y, Kato K, Chung HC, Chen JS, Muro K, Kang WK, Yeh KH, Yoshikawa T, Oh SC, Bai LY, Tamura T, Lee KW, Hamamoto Y, Kim JG, Chin K, Oh DY, Minashi K, Cho JY, Tsuda M, Nishiyama T, Chen LT, Kang YK. Nivolumab in previously treated advanced gastric cancer (ATTRACTION-2): 3-year update and outcome of treatment beyond progression with nivolumab. Gastric Cancer 2021; 24:946-958. [PMID: 33743112 PMCID: PMC8205916 DOI: 10.1007/s10120-021-01173-w] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND ATTRACTION-2 demonstrated that nivolumab improved overall survival (OS) vs placebo in patients with advanced gastric cancer treated with ≥ 2 chemotherapy regimens. However, its long-term efficacy and outcome of treatment beyond progression (TBP) with nivolumab have not been clarified. METHODS The 3-year follow-up data were collected. A subset analysis was performed to explore the efficacy of TBP by assessing postprogression survival (PPS) after the first event of disease progression. RESULTS Overall, 493 patients were randomized (2:1) to receive nivolumab (n = 330) or placebo (n = 163). With a median follow-up of 38.5 (range 36.1-47.5) months, OS of the nivolumab group was significantly longer compared to the placebo group (median 5.3 vs 4.1 months; 3-year survival rate, 5.6% vs 1.9%; hazard ratio [HR], 0.62 [95% confidence interval (CI) 0.50-0.75], P < 0.0001). The median OS of responders (n = 32) who achieved complete response or partial response was 26.7 months and the 3-year survival rate was 35.5% in the nivolumab group. Overall, 109 patients in the nivolumab group and 37 patients in the placebo group received TBP. PPS tended to be longer in the nivolumab group vs placebo group (median 5.8 vs 4.5 months; HR [95% CI], 0.69 [0.47-1.01], P = 0.057). In contrast, PPS was similar between both treatment groups in non-TBP patients (median 2.3 vs 2.2 months; HR 0.90, P = 0.42). CONCLUSIONS Long-term efficacy of nivolumab was confirmed at the 3-year follow-up, and a survival benefit of TBP with nivolumab was suggested. Biomarkers for selecting patients suitable for TBP with nivolumab should be identified in the future.
Collapse
Affiliation(s)
- Narikazu Boku
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Taroh Satoh
- Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, Suita, Japan
| | - Min-Hee Ryu
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ken Kato
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Hyun Cheol Chung
- Division of Medical Oncology, Yonsei Cancer Center, Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Yonsei University Health System, Seoul, South Korea
| | - Jen-Shi Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
, Seoul, South Korea
| | - Kun-Huei Yeh
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan ,Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan ,Present Address: Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Sang Cheul Oh
- Division of Hematology and Oncology, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Takao Tamura
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osakasayama, Japan ,Present Address: Department of Medical Oncology, Kindai University Nara Hospital, Ikoma, Japan
| | - Keun-Wook Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Jong Gwang Kim
- Kyungpook National University School of Medicine, Daegu, South Korea
| | - Keisho Chin
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine
, Seoul, South Korea
| | - Keiko Minashi
- Clinical Trial Promotion Department, Chiba Cancer Center, Chiba, Japan
| | - Jae Yong Cho
- Department of Medical Oncology, Gangnam Severance Hospital, Yonsei University College of Medicine
, Seoul, South Korea
| | - Masahiro Tsuda
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Taihei Nishiyama
- Medical Information, Medical Affairs, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan ,National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan ,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yoon-Koo Kang
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| |
Collapse
|
836
|
Abstract
Introduction: Cancers of the biliary tract (BTC) are aggressive malignancies with limited treatment options and an overall dismal prognosis. In recent years, two concepts, namely precision oncology and immune oncology (IO) have profoundly influenced and, in some cancers, even revolutionized tumor treatments. While positive data from randomized trials have led to the incorporation of targeted concepts for genetically select BTC patients, IO is not yet implemented in clinical practice.Areas covered: We discuss published results from completed, as well as from ongoing studies on IO in BTC, based on a literature search on Pubmed and information provided by clinicaltrials.gov in October 2020. Apart from monotherapy, we outline IO-based combination approaches and highlight pivotal studies whose results will likely influence the future development of relevant concepts in BTC.Expert opinion: Despite partially positive signals, IO thus far disappointed in unselected BTC populations and should currently not be considered as a preferred systemic treatment in patients with microsatellite stable disease outside of clinical trials. In the coming years, a better understanding of the molecular mechanisms underlying resistance to checkpoint inhibition, and the identification of positive predictive biomarkers will be important for the successful integration of IO into treatment concepts for BTC patients.
Collapse
|
837
|
Karstens KF, Stüben BO, Reeh M. Oesophageal Adenocarcinomas: Where Do We Stand Today? Cancers (Basel) 2020; 13:cancers13010109. [PMID: 33396388 PMCID: PMC7796319 DOI: 10.3390/cancers13010109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
|
838
|
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
|
839
|
Slagter AE, Vollebergh MA, Jansen EPM, van Sandick JW, Cats A, van Grieken NCT, Verheij M. Towards Personalization in the Curative Treatment of Gastric Cancer. Front Oncol 2020; 10:614907. [PMID: 33330111 PMCID: PMC7734340 DOI: 10.3389/fonc.2020.614907] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is the fifth most common cancer worldwide and has a high mortality rate. In the last decades, treatment strategy has shifted from an exclusive surgical approach to a multidisciplinary strategy. Treatment options for patients with resectable gastric cancer as recommended by different worldwide guidelines, include perioperative chemotherapy, pre- or postoperative chemoradiotherapy and postoperative chemotherapy. Although gastric cancer is a heterogeneous disease with respect to patient-, tumor-, and molecular characteristics, the current standard of care is still according to a one-size-fits-all approach. In this review, we discuss the background of the different treatment strategies in resectable gastric cancer including the current standard, the specific role of radiotherapy, and describe the current areas of research and potential strategies for personalization of therapy.
Collapse
Affiliation(s)
- Astrid E Slagter
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Marieke A Vollebergh
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Edwin P M Jansen
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Annemieke Cats
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Marcel Verheij
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
840
|
Catenacci DVT, Moya S, Lomnicki S, Chase LM, Peterson BF, Reizine N, Alpert L, Setia N, Xiao SY, Hart J, Siddiqui UD, Hogarth DK, Eng OS, Turaga K, Roggin K, Posner MC, Chang P, Narula S, Rampurwala M, Ji Y, Karrison T, Liao CY, Polite BN, Kindler HL. Personalized Antibodies for Gastroesophageal Adenocarcinoma (PANGEA): A Phase II Study Evaluating an Individualized Treatment Strategy for Metastatic Disease. Cancer Discov 2020; 11:308-325. [PMID: 33234578 DOI: 10.1158/2159-8290.cd-20-1408] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/01/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
The one-year and median overall survival (mOS) rates of advanced gastroesophageal adenocarcinomas (GEA) are ∼50% and <12 months, respectively. Baseline spatial and temporal molecular heterogeneity of targetable alterations may be a cause of failure of targeted/immunooncologic therapies. This heterogeneity, coupled with infrequent incidence of some biomarkers, has resulted in stalled therapeutic progress. We hypothesized that a personalized treatment strategy, applied at first diagnosis then serially over up to three treatment lines using monoclonal antibodies combined with optimally sequenced chemotherapy, could contend with these hurdles. This was tested using a novel clinical expansion-platform type II design with a survival primary endpoint. Of 68 patients by intention-to-treat, the one-year survival rate was 66% and mOS was 15.7 months, meeting the primary efficacy endpoint (one-sided P = 0.0024). First-line response rate (74%), disease control rate (99%), and median progression-free survival (8.2 months) were superior to historical controls. The PANGEA strategy led to improved outcomes warranting a larger randomized study. SIGNIFICANCE: This study highlights excellent outcomes achieved by individually optimizing chemotherapy, biomarker profiling, and matching of targeted therapies at baseline and over time for GEA. Testing a predefined treatment strategy resulted in improved outcomes versus historical controls. Therapeutic resistance observed in correlative analyses suggests that dual targeted inhibition may be beneficial.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois.
| | - Stephanie Moya
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Samantha Lomnicki
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Leah M Chase
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Bryan F Peterson
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Natalie Reizine
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Lindsay Alpert
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Namrata Setia
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Shu-Yuan Xiao
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - John Hart
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Uzma D Siddiqui
- The University of Chicago, Department of Medicine, Center for Endoscopic Research and Therapeutics (CERT), Chicago, Illinois
| | - D Kyle Hogarth
- The University of Chicago, Department of Medicine, Section of Pulmonology, Chicago, Illinois
| | - Oliver S Eng
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | - Kiran Turaga
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | - Kevin Roggin
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | | | - Paul Chang
- The University of Chicago, Department of Radiology, Chicago, Illinois
| | | | | | - Yuan Ji
- The University of Chicago, Department of Public Health Sciences, Chicago, Illinois
| | - Theodore Karrison
- The University of Chicago, Department of Public Health Sciences, Chicago, Illinois
| | - Chih-Yi Liao
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Blase N Polite
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Hedy L Kindler
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| |
Collapse
|
841
|
Emancipator K. Keytruda and PD-L1: a Real-World Example of Co-development of a Drug with a Predictive Biomarker. AAPS JOURNAL 2020; 23:5. [PMID: 33222057 DOI: 10.1208/s12248-020-00525-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022]
Abstract
Pembrolizumab, an anti-programmed death 1 monoclonal antibody, underwent a rapid clinical development program for the treatment of patients with malignancies in parallel to the development of a companion diagnostic for detecting programmed death ligand 1 (PD-L1) expression, which was considered to be a likely biomarker for response and cancer outcome. When investigating outcomes in patients with non-small cell lung cancer (NSCLC) treated with pembrolizumab, PD-L1 tumor proportion score (TPS) was initially identified as a marker of clinical efficacy, with a cutoff of ≥ 50%. This threshold was later reduced to TPS ≥ 1%, which demonstrated that pembrolizumab offered a clinical benefit in a broader population of patients with PD-L1-expressing tumors. As new evidence emerged, it also became clear that immune cell PD-L1 expression has an important role in predicting tumor response in patients treated with pembrolizumab; the combined positive score (CPS), which accounts for both tumor and immune cell PD-L1 expression, was then applied in clinical studies for indications outside of NSCLC. This review summarizes the outcomes of landmark studies in the KEYNOTE clinical development program that investigated the efficacy of pembrolizumab in patients with a variety of malignancies, the relative outcomes when patients were stratified by PD-L1 status, and how these observations have influenced the approved indications for pembrolizumab.
Collapse
Affiliation(s)
- Kenneth Emancipator
- Department of Translational Medicine, Merck & Co., Inc, 2000 Galloping Hill Road, Kenilworth, New Jersey, 07033, USA.
| |
Collapse
|
842
|
Li W, Li Q, Yu Y, Wang Y, Chen E, Chen L, Wang Z, Cui Y, Liu T. Effect of Immune Checkpoint Inhibitors Plus Chemotherapy on Advanced Gastric Cancer Patients with Elevated Serum AFP or Hepatoid Adenocarcinoma. Cancer Manag Res 2020; 12:11113-11119. [PMID: 33173344 PMCID: PMC7646478 DOI: 10.2147/cmar.s276969] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Alpha-fetoprotein-producing gastric cancer (AFPGC) and hepatoid adenocarcinoma of stomach (HAS) are rare types of gastric cancer, with specific clinical manifestations and poor prognosis. The standardized treatment process of such cancers remains elusive. We aim to investigate the efficacy of immunotherapy combined with chemotherapy on patients with AFPGC or HAS. PATIENTS AND METHODS AFPGC and HAS patients who underwent immunotherapy and/or chemotherapy as the first-line treatment at our institute from June 2016 to December 2018 were enrolled in this observational study. Their clinicopathological characteristics, serum AFP level and treatment methods were collected. The progression-free survival (PFS) and overall survival (OS) were analyzed and compared between patients who received immunotherapy plus chemotherapy and those received chemotherapy. RESULTS A total of 21 patients with advanced AFPGC or HAS were included in the study and the median follow-up time was 28.0 months. Of the 21 patients, 7 patients received immunotherapy of PD-1 antibody (nivolumab) plus chemotherapy and 14 patients as control received chemotherapy with or without Herceptin/Apatinib. The median progression-free survival (mPFS) time was 5.0 months (4.3 months in the control group and 22.0 months in the immunotherapy group). The median overall survival (mOS) time of the control group was 16.0 months (14.0 months in chemotherapy alone subgroup, 20.0 months in chemotherapy plus Apatinib or Herceptin subgroup), while the mOS of patients receiving immunotherapy was not reached. CONCLUSION This study suggested PD-1 checkpoint inhibitor plus chemotherapy could benefit AFPGC and HAS patients. Its mechanism of action warrants further investigation.
Collapse
Affiliation(s)
- Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Erbao Chen
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Lingli Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Zhiming Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Center of Evidence-Based Medicine, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|