30
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Effect of Combined Immune Checkpoint Inhibition vs Best Supportive Care Alone in Patients With Advanced Colorectal Cancer : The Canadian Cancer Trials Group CO.26 Study

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Key Points

          Question

          Can combined immune checkpoint inhibition improve overall survival (OS) in patients with advanced refractory colorectal cancer (CRC)?

          Findings

          In this randomized phase 2 study with 180 patients randomized in a 2:1 ratio to tremelimumab and durvalumab plus best supportive care or best supportive care alone, the median OS was 6.6 months for durvalumab and tremelimumab and 4.1 months for best supportive care; correlative analysis revealed that patients with plasma tumor mutation burden (TMB) of 28 or more variants per megabase had the greatest OS benefit.

          Meaning

          Combined immune checkpoint inhibition may prolong OS in patients with advanced refractory CRC.

          Abstract

          This phase 2 randomized clinical trial evaluates the outcomes of combined immune checkpoint inhibition plus best supportive care vs best supportive care alone for patients with advanced colorectal cancer.

          Abstract

          Importance

          Single-agent immune checkpoint inhibition has not shown activities in advanced refractory colorectal cancer (CRC), other than in those patients who are microsatellite-instability high (MSI-H).

          Objective

          To evaluate whether combining programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte–associated protein 4 (CTLA-4) inhibition improved patient survival in metastatic refractory CRC.

          Design, Setting, and Participants

          A randomized phase 2 study was conducted in 27 cancer centers across Canada between August 2016 and June 2017, and data were analyzed on October 18, 2018. Eligible patients had histologically confirmed adenocarcinoma of the colon or rectum; received all available standard systemic therapies (fluoropyrimidines, oxaliplatin, irinotecan, and bevacizumab if appropriate; cetuximab or panitumumab if RAS wild-type tumors; regorafenib if available); were aged 18 years or older; had adequate organ function; had Eastern Cooperative Oncology Group performance status of 0 or 1, and measurable disease.

          Interventions

          We randomly assigned patients to receive either 75 mg of tremelimumab every 28 days for the first 4 cycles plus 1500 mg durvalumab every 28 days, or best supportive care alone (BSC) in a 2:1 ratio.

          Main Outcomes and Measures

          The primary end point was overall survival (OS) and a 2-sided P<.10 was considered statistically significant. Circulating cell-free DNA from baseline plasma was used to determine microsatellite instability (MSI) and tumor mutation burden (TMB).

          Results

          Of 180 patients enrolled (121 men [67.2%] and 59 women [32.8%]; median [range] age, 65 [36-87] years), 179 were treated. With a median follow-up of 15.2 months, the median OS was 6.6 months for durvalumab and tremelimumab and 4.1 months for BSC (hazard ratio [HR], 0.72; 90% CI, 0.54-0.97; P = .07). Progression-free survival was 1.8 months and 1.9 months respectively (HR, 1.01; 90% CI, 0.76-1.34). Grade 3 or 4 adverse events were significantly more frequent with immunotherapy (75 [64%] patients in the treatment group had at least 1 grade 3 or higher adverse event vs 12 [20%] in the BSC group). Circulating cell-free DNA analysis was successful in 168 of 169 patients with available samples. In patients who were microsatellite stable (MSS), OS was significantly improved with durvalumab and tremelimumab (HR, 0.66; 90% CI, 0.49-0.89; P = .02). Patients who were MSS with plasma TMB of 28 variants per megabase or more (21% of MSS patients) had the greatest OS benefit (HR, 0.34; 90% CI, 0.18-0.63; P = .004).

          Conclusions and Relevance

          This phase 2 study suggests that combined immune checkpoint inhibition with durvalumab plus tremelimumab may be associated with prolonged OS in patients with advanced refractory CRC. Elevated plasma TMB may select patients most likely to benefit from durvalumab and tremelimumab. Further confirmation studies are warranted.

          Trial Registration

          ClinicalTrials.gov Identifier: NCT02870920

          Related collections

          Most cited references12

          • Record: found
          • Abstract: found
          • Article: not found

          Effect of First-Line Chemotherapy Combined With Cetuximab or Bevacizumab on Overall Survival in Patients With KRAS Wild-Type Advanced or Metastatic Colorectal Cancer: A Randomized Clinical Trial.

          Combining biologic monoclonal antibodies with chemotherapeutic cytotoxic drugs provides clinical benefit to patients with advanced or metastatic colorectal cancer, but the optimal choice of the initial biologic therapy in previously untreated patients is unknown.
            • Record: found
            • Abstract: found
            • Article: not found

            Genetic diversity of tumors with mismatch repair deficiency influences anti–PD-1 immunotherapy response

            Tumors with mismatch repair deficiency (MMR-d) are characterized by sequence alterations in microsatellites and can accumulate thousands of mutations. This high mutational burden renders tumors immunogenic and sensitive to programmed cell death–1 (PD-1) immune checkpoint inhibitors. Yet, despite their tumor immunogenicity, patients with MMR-deficient tumors experience highly variable responses, and roughly half are refractory to treatment. We present experimental and clinical evidence showing that the degree of microsatellite instability (MSI) and resultant mutational load, in part, underlies the variable response to PD-1 blockade immunotherapy in MMR-d human and mouse tumors. The extent of response is particularly associated with the accumulation of insertion-deletion (indel) mutational load. This study provides a rationale for the genome-wide characterization of MSI intensity and mutational load to better profile responses to anti–PD-1 immunotherapy across MMR-deficient human cancers.
              • Record: found
              • Abstract: found
              • Article: not found

              Adaptive mutability of colorectal cancers in response to targeted therapies

              The emergence of drug resistance limits the efficacy of targeted therapies in human tumors. The prevalent view is that resistance is a fait accompli: when treatment is initiated, cancers already contain drug-resistant mutant cells. Bacteria exposed to antibiotics transiently increase their mutation rates (adaptive mutability), thus improving the likelihood of survival. We investigated whether human colorectal cancer (CRC) cells likewise exploit adaptive mutability to evade therapeutic pressure. We found that epidermal growth factor receptor (EGFR)/BRAF inhibition down-regulates mismatch repair (MMR) and homologous recombination (HR) DNA repair genes, and concomitantly up-regulates error-prone polymerases in drug-tolerant (persister) cells. MMR proteins were also down-regulated in patient-derived xenografts and tumor specimens during therapy. EGFR/BRAF inhibition induced DNA damage, increased mutability and triggered microsatellite instability. Thus, like unicellular organisms, tumor cells evade therapeutic pressures by enhancing mutability.

                Author and article information

                Journal
                JAMA Oncol
                JAMA Oncol
                JAMA Oncol
                JAMA Oncology
                American Medical Association
                2374-2437
                2374-2445
                June 2020
                7 May 2020
                7 May 2020
                : 6
                : 6
                : 1-8
                Affiliations
                [1 ]Princess Margaret Cancer Center, Toronto, Canada
                [2 ]The Ottawa Hospital, Ottawa, Canada
                [3 ]BC Cancer, Vancouver, Canada
                [4 ]Virginia Mason Medical Center, Seattle
                [5 ]Department of Oncology, Queen’s University, Kingston, Canada
                [6 ]CHU de Québec-Université, Laval, Canada
                [7 ]Eastern Health, St John’s, Canada
                [8 ]Juvravinski Cancer Center, Hamilton, Canada
                [9 ]Segal Cancer Center, Montreal, Canada
                [10 ]Moncton Hospital, Moncton, Canada
                [11 ]Dalhousie University, Halifax, Canada
                [12 ]Hôpital Sacré-Coeur de Montréal, Montreal, Canada
                [13 ]Sherbrooke University, Sherbrooke, Canada
                [14 ]Saskatoon Cancer Center, Saskatoon, Canada
                [15 ]Hôpital Cité-de-la-Santé, Laval, Canada
                [16 ]Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Canada
                [17 ]Cross Cancer Center, Edmonton, Canada
                [18 ]Canadian Cancer Trials Group, Kingston, Canada
                Author notes
                Article Information
                Corresponding Author: Eric X. Chen, MD, PhD, Princess Margaret Cancer Center, 700 University Ave, Rm 7-824, Toronto, ON M5G 1X6 Canada ( eric.chen@ 123456uhn.ca ).
                Accepted for Publication: March 2, 2020.
                Published Online: May 7, 2020. doi:10.1001/jamaoncol.2020.0910
                Open Access: This is an open access article distributed under the terms of the CC-BY License. © 2020 Chen EX et al. JAMA Oncology.
                Author Contributions: Drs Chen and O’Callaghan had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.
                Concept and design: Chen, Jonker, Loree, Ahmad, Kavan, Tu, O'Callaghan.
                Acquisition, analysis, or interpretation of data: Chen, Jonker, Loree, Kennecke, Berry, Couture, Goffin, Kavan, Harb, Colwell, Samimi, Samson, Abbas, Aucoin, Aubin, Koski, Wei, Magoski, Tu, O'Callaghan.
                Drafting of the manuscript: Chen, Jonker, Loree, Ahmad, Kavan, Abbas.
                Critical revision of the manuscript for important intellectual content: Chen, Jonker, Loree, Kennecke, Berry, Couture, Goffin, Kavan, Harb, Colwell, Samimi, Samson, Aucoin, Aubin, Koski, Wei, Magoski, Tu, O'Callaghan .
                Statistical analysis: Loree, Tu, O'Callaghan.
                Obtained funding: Jonker, Aucoin, O'Callaghan.
                Administrative, technical, or material support: Chen, Jonker, Loree, Kennecke, Couture, Goffin, Samimi, Aucoin, Aubin, Wei, Magoski, O'Callaghan.
                Supervision: Jonker, Ahmad, Kavan, Samimi, Abbas, Aubin, Koski, O'Callaghan .
                Conflict of Interest Disclosures: Dr Chen participated in clinical trials sponsored by Merck, BMS, Boston Biomedical, AZ, and received research support (drugs only) from AZ. Dr Chen received honoraria from Eisai and Taiho. Dr Loree received research support from Ipsen and honoraria from Ipsen, Amgen, Bayer, Novartis, and Taiho. Dr Samson received honorarium from Taiho. Dr Goffin received honoraria from Merck and Amgen and nonfinancial support from AZ. Dr Samimi received honoraria from Amgen, Celgene, Ipsen, Apobiologix, Eisai, Taiho, and Roche. Dr Aubin received honoraria from Taiho, Shire, Amgen, BMS, and Celgene, and participated in clinical trials sponsored by Merck and BMS. Dr Aucoin received honoraria from AZ, Roche, BMS, Amgen, Takeda, Pfizer, Taiho, Celgene. Dr Wei received honoraria from Shire and Celgene, and nonfinancial support from Bayer. All other authors declared no competing interests.
                Data Sharing Statement: See Supplement 3.
                Funding/Support: AstraZeneca provided durvalumab and tremelimumab and contributed partial funding for this study. The Canadian Cancer Trials Group (CCTG) is funded by the Canadian Cancer Society.
                Role of the Funder/Sponsor: AstraZeneca was consulted on design of the study, and reviewed a draft of the manuscript. It played no role in conduct of the study; collection, management, analysis, and interpretation of the data; preparation, or approval of the manuscript; and decision to submit the manuscript for publication.
                Meeting Presentation: Presented at ASCO GI Symposium, January 19, 2019, San Francisco, California; and at ASCO Annual Meeting, June 3, 2019, Chicago, Illinois.
                Article
                coi200014
                10.1001/jamaoncol.2020.0910
                7206536
                32379280
                9effbfaa-b8d6-4b88-a334-001eaaab7d4d
                Copyright 2020 Chen EX et al. JAMA Oncology.

                This is an open access article distributed under the terms of the CC-BY License.

                History
                : 4 December 2019
                : 2 March 2020
                Categories
                Research
                Research
                Original Investigation
                Featured
                Online First
                Comments

                Comments

                Comment on this article

                Related Documents Log