101
|
Cohen R, Shi Q, André T. Immunotherapy for Early Stage Colorectal Cancer: A Glance into the Future. Cancers (Basel) 2020; 12:E1990. [PMID: 32708216 PMCID: PMC7409300 DOI: 10.3390/cancers12071990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) have reshaped therapeutic strategies for cancer patients. The development of ICI for early stage colorectal cancer is accompanied by specific challenges: (i) the selection of patients who are likely to benefit from these treatments, i.e., patients with tumors harboring predictive factors of efficacy of ICI, such as microsatellite instability and/or mismatch repair deficiency (MSI/dMMR), or other potential parameters (increased T cell infiltration using Immunoscore® or others, high tumor mutational burden, POLE mutation), (ii) the selection of patients at risk of disease recurrence (poor prognostic features), and (iii) the choice of an accurate clinical trial methodological framework. In this review, we will discuss the ins and outs of clinical research of ICI for early stage MSI/dMMR CC patients in adjuvant and neoadjuvant settings. We will then summarize data that might support the development of ICI in localized colorectal cancer beyond MSI/dMMR.
Collapse
Affiliation(s)
- Romain Cohen
- Department of Medical Oncology, Hôpital Saint-Antoine, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), F-75012 Paris, France;
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA;
| | - Qian Shi
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA;
| | - Thierry André
- Department of Medical Oncology, Hôpital Saint-Antoine, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), F-75012 Paris, France;
| |
Collapse
|
102
|
Kawazoe A, Kuboki Y, Shinozaki E, Hara H, Nishina T, Komatsu Y, Yuki S, Wakabayashi M, Nomura S, Sato A, Kuwata T, Kawazu M, Mano H, Togashi Y, Nishikawa H, Yoshino T. Multicenter Phase I/II Trial of Napabucasin and Pembrolizumab in Patients with Metastatic Colorectal Cancer (EPOC1503/SCOOP Trial). Clin Cancer Res 2020; 26:5887-5894. [PMID: 32694160 DOI: 10.1158/1078-0432.ccr-20-1803] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/18/2020] [Accepted: 07/15/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE This is a phase I/II trial to assess the efficacy and safety of napabucasin plus pembrolizumab for metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Phase I was conducted to determine the recommended phase 2 dose (RP2D) in a dose escalation design of napabucasin (240 to 480 mg twice daily) with 200 mg pembrolizumab every 3 weeks. Phase II included cohort A (n = 10, microsatellite instability high, MSI-H) and cohort B (n = 40, microsatellite stable, MSS). The primary endpoint was immune-related objective response rate (irORR). PD-L1 combined positive score (CPS), genomic profiles, and the consensus molecular subtypes (CMS) of colorectal cancer were assessed. RESULTS A total of 55 patients were enrolled in this study. In phase I, no patients experienced dose-limiting toxicities, and napabucasin 480 mg was determined as RP2D. The irORR was 50.0% in cohort A and 10.0% in cohort B. In cohort B, the irORR was 0%, 5.3%, and 42.9% in CPS < 1, 1≤ CPS <10, and CPS ≥ 10, respectively. Patients with objective response tended to have higher tumor mutation burden than those without. Of evaluable 18 patients for CMS classification in cohort B, the irORR was 33.3%, 0%, 33.3%, and 33.3% in CMS1, CMS2, CMS3, and CMS4, respectively. The common grade 3 or higher treatment-related adverse events included fever (10.0%) in cohort A and decreased appetite (7.5%) and diarrhea (5.0%) in cohort B. CONCLUSIONS Napabucasin with pembrolizumab showed antitumor activity with acceptable toxicities for patients with MSS mCRC as well as MSI-H mCRC, although it did not meet the primary end point. The impact of related biomarkers on the efficacy warrants further investigations in the additional cohort.See related commentary by Nusrat, p. 5775.
Collapse
Affiliation(s)
- Akihito Kawazoe
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yasutoshi Kuboki
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Eiji Shinozaki
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, Shikoku Cancer Center, Matsuyama, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Hokkaido, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Masashi Wakabayashi
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Shogo Nomura
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Takeshi Kuwata
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, National Cancer Center, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center, Tokyo, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Hospital East, Chiba, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Hospital East, Chiba, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
103
|
Lai WA, Liu CY, Lin SY, Chen CC, Hang JF. Characterization of Driver Mutations in Anaplastic Thyroid Carcinoma Identifies RAS and PIK3CA Mutations as Negative Survival Predictors. Cancers (Basel) 2020; 12:E1973. [PMID: 32698386 PMCID: PMC7409295 DOI: 10.3390/cancers12071973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/18/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is rare but highly aggressive. We investigated the association of selected driver mutations, including BRAF, RAS, PIK3CA, TERT promoter, TP53, POLE, and mismatch repair deficiency (MMR-D) with the clinicopathological features of ATC to identify prognostic and predictive biomarkers. Thirty-nine retrospective cases from pathology archives were enrolled for clinicopathology analysis and immunohistochemistry, and 27 cases had sufficient specimens for further molecular testing using targeted next-generation sequencing and mass spectrometry. BRAFV600E and RAS mutations were identified in 25.9% and 40.7% of ATC, respectively. BRAFV600E mutation was significantly associated with coexisting papillary thyroid carcinoma (p = 0.009) and RAS mutations with female gender (p = 0.012). In univariant analysis, the non-BRAF/RAS tumors were significantly associated with the presence of a sarcomatoid pattern (p = 0.045). PIK3CA, TERT promoter, and TP53 mutations were identified in 14.8%, 81.5%, and 70.4% of cases, respectively. No MMR-D or POLE mutations were detected. In survival analyses, RAS and PIK3CA mutations were significantly associated with inferior outcomes (p = 0.03 and p = 0.006, respectively). In conclusion, driver mutations in ATC are associated with distinct clinicopathological features. RAS and PIK3CA mutations were negative predictors for patient survival. Emerging therapeutic agents targeting BRAF, RAS, and PI3 kinase may benefit a substantial proportion of ATC patients.
Collapse
Affiliation(s)
- Wei-An Lai
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-A.L.); (S.-Y.L.)
| | - Chih-Yi Liu
- Division of Pathology, Sijhih Cathay General Hospital, New Taipei City 221, Taiwan;
| | - Shih-Yao Lin
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-A.L.); (S.-Y.L.)
- Department of Pathology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan;
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Jen-Fan Hang
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (W.-A.L.); (S.-Y.L.)
- National Yang-Ming University School of Medicine, Taipei 112, Taiwan
| |
Collapse
|
104
|
Millen R, Hendry S, Narasimhan V, Abbott R, Croxford M, Gibbs P, Tie J, Wong H, Jones I, Kosmider S, Byrne D, Zalcberg J, Fox S, Desai J, Visvanathan K, Ramsay RG, Tran B. CD8 + tumor-infiltrating lymphocytes within the primary tumor of patients with synchronous de novo metastatic colorectal carcinoma do not track with survival. Clin Transl Immunology 2020; 9:e1155. [PMID: 32953115 PMCID: PMC7484874 DOI: 10.1002/cti2.1155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Tumor-infiltrating lymphocytes (TIL), particularly CD8+ TILs in patients with colorectal cancer (CRC), are highly prognostic in the early-disease stages (I-II). In metastatic disease (stage IV; mCRC), their influence is less well defined. It has presumably failed to contain tumor cells to the primary site; however, is this evident? We explored the prognostic impact of TILs at the primary site in patients who presented de novo with mCRC. METHODS Treatment-naïve patients (109) with mCRC were assessed for CD8+ TILs and PD-L1 expression. Microsatellite instability (MSI) was evaluated by IHC for PMS2 and MSH6 proteins and/or by PCR using the Bethesda panel. RESULTS Microsatellite instability-high tumors had significantly more CD8+ TILs, with no significant survival advantage observed between MSI-H and microsatellite stable (MSS) tumors (12 vs 19 months, P = 0.304). TIL density for all cases had no impact on OS (low: 20 vs high: 13 months, P = 0.426), while PD-L1 of 1% or higher was associated with reduced mean survival (9.6 vs 18.9 months; P = 0.038). MSI-H tumors and associated immune cells had higher PD-L1 expression than in MSS cases. A positive correlation between PD-L1 on immune cells and CD8+ve TILs was found. A subset of MSS tumors had relatively high TILs approximating that of MSI-H tumors. CONCLUSION In contrast to early-stage CRC, the immune response in primary tumors of patients with de novo mCRC does not appear to influence survival. A subgroup of MSS tumors was identified with increased TILs/PD-L1 comparable to MSI-H tumors, traditionally not be considered for immune checkpoint blockade and perhaps should be.
Collapse
Affiliation(s)
- Rosemary Millen
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
- St. Vincent's HospitalMelbourneVICAustralia
| | - Shona Hendry
- Department of Anatomical PathologySt Vincent's Hospital MelbourneMelbourneVICAustralia
- Department of PathologyUniversity of MelbourneMelbourneVICAustralia
| | - Vignesh Narasimhan
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | - Rebecca Abbott
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | | | - Peter Gibbs
- Royal Melbourne HospitalMelbourneVICAustralia
- Walter and Eliza Hall InstituteParkvilleVICAustralia
| | - Jeanne Tie
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
- Western HealthFootscrayVICAustralia
- Royal Melbourne HospitalMelbourneVICAustralia
- Walter and Eliza Hall InstituteParkvilleVICAustralia
| | - Hui‐Li Wong
- Royal Melbourne HospitalMelbourneVICAustralia
- Walter and Eliza Hall InstituteParkvilleVICAustralia
| | - Ian Jones
- Royal Melbourne HospitalMelbourneVICAustralia
| | - Suzanne Kosmider
- Western HealthFootscrayVICAustralia
- Royal Melbourne HospitalMelbourneVICAustralia
| | - David Byrne
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | - John Zalcberg
- Monash UniversityMelbourneVICAustralia
- Alfred HealthPrahranVICAustralia
| | - Stephen Fox
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | - Jayesh Desai
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | - Kumar Visvanathan
- St. Vincent's HospitalMelbourneVICAustralia
- University of MelbourneMelbourneVICAustralia
| | - Robert G Ramsay
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | - Ben Tran
- Peter MacCallum Cancer CentreMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
- Royal Melbourne HospitalMelbourneVICAustralia
- Walter and Eliza Hall InstituteParkvilleVICAustralia
| |
Collapse
|
105
|
Siraj AK, Bu R, Iqbal K, Parvathareddy SK, Masoodi T, Siraj N, Al-Rasheed M, Kong Y, Ahmed SO, Al-Obaisi KAS, Victoria IG, Arshad M, Al-Dayel F, Abduljabbar A, Ashari LH, Al-Kuraya KS. POLE and POLD1 germline exonuclease domain pathogenic variants, a rare event in colorectal cancer from the Middle East. Mol Genet Genomic Med 2020; 8:e1368. [PMID: 32567205 PMCID: PMC7434734 DOI: 10.1002/mgg3.1368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Colorectal cancer (CRC) is a major contributor to morbidity and mortality related to cancer. Only ~5% of all CRCs occur as a result of pathogenic variants in well‐defined CRC predisposing genes. The frequency and effect of exonuclease domain pathogenic variants of POLE and POLD1 genes in Middle Eastern CRCs is still unknown. Methods Targeted capture sequencing and Sanger sequencing technologies were employed to investigate the germline exonuclease domain pathogenic variants of POLE and POLD1 in Middle Eastern CRCs. Immunohistochemical analysis of POLE and POLD1 was performed to look for associations between protein expression and clinico‐pathological characteristics. Results Five damaging or possibly damaging variants (0.44%) were detected in 1,135 CRC cases, four in POLE gene (0.35%, 4/1,135) and one (0.1%, 1/1,135) in POLD1 gene. Furthermore, low POLE protein expression was identified in 38.9% (417/1071) cases and a significant association with lymph node involvement (p = .0184) and grade 3 tumors (p = .0139) was observed. Whereas, low POLD1 expression was observed in 51.9% (555/1069) of cases and was significantly associated with adenocarcinoma histology (p = .0164), larger tumor size (T3 and T4 tumors; p = .0012), and stage III tumors (p = .0341). Conclusion POLE and POLD1 exonuclease domain pathogenic variants frequency in CRC cases was very low and these exonuclease domain pathogenic variants might be rare causative events of CRC in the Middle East. POLE and POLD1 can be included in multi‐gene panels to screen CRC patients.
Collapse
Affiliation(s)
- Abdul K Siraj
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Rong Bu
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Kaleem Iqbal
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Sandeep K Parvathareddy
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Tariq Masoodi
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Nabil Siraj
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Maha Al-Rasheed
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Yan Kong
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Saeeda O Ahmed
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Khadija A S Al-Obaisi
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Ingrid G Victoria
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Maham Arshad
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| | - Fouad Al-Dayel
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alaa Abduljabbar
- Colorectal Section, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Luai H Ashari
- Colorectal Section, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Khawla S Al-Kuraya
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, iyadh, Saudi Arabia
| |
Collapse
|
106
|
Onuma AE, Zhang H, Huang H, Williams TM, Noonan A, Tsung A. Immune Checkpoint Inhibitors in Hepatocellular Cancer: Current Understanding on Mechanisms of Resistance and Biomarkers of Response to Treatment. Gene Expr 2020; 20:53-65. [PMID: 32340652 PMCID: PMC7284108 DOI: 10.3727/105221620x15880179864121] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy worldwide and a leading cause of death worldwide. Its incidence continues to increase in the US due to hepatitis C infection and nonalcoholic steatohepatitis. Liver transplantation and resection remain the best therapeutic options for cure, but these are limited by the shortage of available organs for transplantation, diagnosis at advanced stage, and underlying chronic liver disease found in most patients with HCC. Immune checkpoint inhibitors (ICIs) have been shown to be an evolving novel treatment option in certain advanced solid tumors and have been recently approved for inoperable, advanced, and metastatic HCC. Unfortunately, a large cohort of patients with HCC fail to respond to immunotherapy. In this review, we discuss the ICIs currently approved for HCC treatment and their various mechanisms of action. We will highlight current understanding of mechanism of resistance and limitations to ICIs. Finally, we will describe emerging biomarkers of response to ICIs and address future direction on overcoming resistance to immune checkpoint therapy.
Collapse
Affiliation(s)
- Amblessed E. Onuma
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hongji Zhang
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- †Department of Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hai Huang
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Terence M. Williams
- ‡Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anne Noonan
- §Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Allan Tsung
- *Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
107
|
Caracciolo D, Riillo C, Arbitrio M, Di Martino MT, Tagliaferri P, Tassone P. Error-prone DNA repair pathways as determinants of immunotherapy activity: an emerging scenario for cancer treatment. Int J Cancer 2020; 147:2658-2668. [PMID: 32383203 DOI: 10.1002/ijc.33038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/18/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
Defects in DNA repair machinery play a critical role in the pathogenesis and progression of human cancer. When they occur, the tumor cells activate error-prone mechanisms which lead to genomic instability and high mutation rate. These defects represent, therefore, a cancer Achilles'heel which could be therapeutically exploited by the use of DNA damage response inhibitors. Moreover, experimental and clinical evidence indicates that DNA repair deregulation has a pivotal role also in promoting immune recognition and immune destruction of cancer cells. Indeed, immune checkpoint inhibitors have received regulatory approval in tumors characterized by high genomic instability, such as melanomas and lung cancer. Here, we discuss how deregulation of DNA repair, through activation of error-prone mechanisms, increases immune activation against cancer. Finally, we address the potential strategies to use DNA repair components as biomarkers and/or therapeutic targets to empower immune-oncology treatment of human cancer.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | | | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
108
|
Tsimberidou AM, Fountzilas E, Nikanjam M, Kurzrock R. Review of precision cancer medicine: Evolution of the treatment paradigm. Cancer Treat Rev 2020; 86:102019. [PMID: 32251926 PMCID: PMC7272286 DOI: 10.1016/j.ctrv.2020.102019] [Citation(s) in RCA: 392] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
In recent years, biotechnological breakthroughs have led to identification of complex and unique biologic features associated with carcinogenesis. Tumor and cell-free DNA profiling, immune markers, and proteomic and RNA analyses are used to identify these characteristics for optimization of anticancer therapy in individual patients. Consequently, clinical trials have evolved, shifting from tumor type-centered to gene-directed, histology-agnostic, with innovative adaptive design tailored to biomarker profiling with the goal to improve treatment outcomes. A plethora of precision medicine trials have been conducted. The majority of these trials demonstrated that matched therapy is associated with superior outcomes compared to non-matched therapy across tumor types and in specific cancers. To improve the implementation of precision medicine, this approach should be used early in the course of the disease, and patients should have complete tumor profiling and access to effective matched therapy. To overcome the complexity of tumor biology, clinical trials with combinations of gene-targeted therapy with immune-targeted approaches (e.g., checkpoint blockade, personalized vaccines and/or chimeric antigen receptor T-cells), hormonal therapy, chemotherapy and/or novel agents should be considered. These studies should target dynamic changes in tumor biologic abnormalities, eliminating minimal residual disease, and eradicating significant subclones that confer resistance to treatment. Mining and expansion of real-world data, facilitated by the use of advanced computer data processing capabilities, may contribute to validation of information to predict new applications for medicines. In this review, we summarize the clinical trials and discuss challenges and opportunities to accelerate the implementation of precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX.
| | - Elena Fountzilas
- Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece
| | - Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| |
Collapse
|
109
|
Lai E, Liscia N, Donisi C, Mariani S, Tolu S, Pretta A, Persano M, Pinna G, Balconi F, Pireddu A, Impera V, Dubois M, Migliari M, Spanu D, Saba G, Camera S, Musio F, Ziranu P, Puzzoni M, Demurtas L, Pusceddu V, Dettori M, Massa E, Atzori F, Dessì M, Astara G, Madeddu C, Scartozzi M. Molecular-Biology-Driven Treatment for Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:E1214. [PMID: 32413973 PMCID: PMC7281737 DOI: 10.3390/cancers12051214] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Metastatic CRC (mCRC) is a molecular heterogeneous disease. The aim of this review is to give an overview of molecular-driven treatment of mCRC patients. METHODS A review of clinical trials, retrospective studies and case reports was performed regarding molecular biomarkers with therapeutic implications. RESULTS RAS wild-type status was confirmed as being crucial for anti-epidermal growth factor receptor (EGFR) monoclonal antibodies and for rechallenge strategy. Antiangiogenic therapies improve survival in first- and second-line settings, irrespective of RAS status, while tyrosine kinase inhibitors (TKIs) remain promising in refractory mCRC. Promising results emerged from anti-HER2 drugs trials in HER2-positive mCRC. Target inhibitors were successful for BRAFV600E mutant mCRC patients, while immunotherapy was successful for microsatellite instability-high/defective mismatch repair (MSI-H/dMMR) or DNA polymerase epsilon catalytic subunit (POLE-1) mutant patients. Data are still lacking on NTRK, RET, MGMT, and TGF-β, which require further research. CONCLUSION Several molecular biomarkers have been identified for the tailored treatment of mCRC patients and multiple efforts are currently ongoing to increase the therapeutic options. In the era of precision medicine, molecular-biology-driven treatment is the key to impro patient selection and patient outcomes. Further research and large phase III trials are required to ameliorate the therapeutic management of these patients.
Collapse
Affiliation(s)
- Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Nicole Liscia
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Simona Tolu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Giovanna Pinna
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Francesca Balconi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Annagrazia Pireddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Valentino Impera
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Dubois
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Marco Migliari
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Silvia Camera
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesca Musio
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Laura Demurtas
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Manuela Dettori
- Medical Oncology Unit, Azienda Ospedaliera Brotzu, Ospedale Businco, 09134 Cagliari, Italy
| | - Elena Massa
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Francesco Atzori
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Mariele Dessì
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Giorgio Astara
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Clelia Madeddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| |
Collapse
|
110
|
Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther 2020; 5:22. [PMID: 32296018 PMCID: PMC7082344 DOI: 10.1038/s41392-020-0116-z] [Citation(s) in RCA: 997] [Impact Index Per Article: 199.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is among the most lethal and prevalent malignancies in the world and was responsible for nearly 881,000 cancer-related deaths in 2018. Surgery and chemotherapy have long been the first choices for cancer patients. However, the prognosis of CRC has never been satisfying, especially for patients with metastatic lesions. Targeted therapy is a new optional approach that has successfully prolonged overall survival for CRC patients. Following successes with the anti-EGFR (epidermal growth factor receptor) agent cetuximab and the anti-angiogenesis agent bevacizumab, new agents blocking different critical pathways as well as immune checkpoints are emerging at an unprecedented rate. Guidelines worldwide are currently updating the recommended targeted drugs on the basis of the increasing number of high-quality clinical trials. This review provides an overview of existing CRC-targeted agents and their underlying mechanisms, as well as a discussion of their limitations and future trends.
Collapse
Affiliation(s)
- Yuan-Hong Xie
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China.
| |
Collapse
|
111
|
Wang Y, Zhang X, Xiao X, Zhang FR, Yan X, Feng X, Zhao Z, Guan Y, Wang J. Accurately estimating the length distributions of genomic micro-satellites by tumor purity deconvolution. BMC Bioinformatics 2020; 21:82. [PMID: 32164528 PMCID: PMC7069170 DOI: 10.1186/s12859-020-3349-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Genomic micro-satellites are the genomic regions that consist of short and repetitive DNA motifs. Estimating the length distribution and state of a micro-satellite region is an important computational step in cancer sequencing data pipelines, which is suggested to facilitate the downstream analysis and clinical decision supporting. Although several state-of-the-art approaches have been proposed to identify micro-satellite instability (MSI) events, they are limited in dealing with regions longer than one read length. Moreover, based on our best knowledge, all of these approaches imply a hypothesis that the tumor purity of the sequenced samples is sufficiently high, which is inconsistent with the reality, leading the inferred length distribution to dilute the data signal and introducing the false positive errors. RESULTS In this article, we proposed a computational approach, named ELMSI, which detected MSI events based on the next generation sequencing technology. ELMSI can estimate the specific length distributions and states of micro-satellite regions from a mixed tumor sample paired with a control one. It first estimated the purity of the tumor sample based on the read counts of the filtered SNVs loci. Then, the algorithm identified the length distributions and the states of short micro-satellites by adding the Maximum Likelihood Estimation (MLE) step to the existing algorithm. After that, ELMSI continued to infer the length distributions of long micro-satellites by incorporating a simplified Expectation Maximization (EM) algorithm with central limit theorem, and then used statistical tests to output the states of these micro-satellites. Based on our experimental results, ELMSI was able to handle micro-satellites with lengths ranging from shorter than one read length to 10kbps. CONCLUSIONS To verify the reliability of our algorithm, we first compared the ability of classifying the shorter micro-satellites from the mixed samples with the existing algorithm MSIsensor. Meanwhile, we varied the number of micro-satellite regions, the read length and the sequencing coverage to separately test the performance of ELMSI on estimating the longer ones from the mixed samples. ELMSI performed well on mixed samples, and thus ELMSI was of great value for improving the recognition effect of micro-satellite regions and supporting clinical decision supporting. The source codes have been uploaded and maintained at https://github.com/YixuanWang1120/ELMSI for academic use only.
Collapse
Affiliation(s)
- Yixuan Wang
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| | - Xuanping Zhang
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| | - Xiao Xiao
- Institute of Health Administration and Policy, School of Public Policy and Administration, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| | - Fei-Ran Zhang
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong People’s Republic of China
| | - Xinxing Yan
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| | - Xuan Feng
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| | - Zhongmeng Zhao
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| | - Yanfang Guan
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Geneplus Beijing Institute, Beijing, 100061 People’s Republic of China
| | - Jiayin Wang
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
- Shaanxi Engineering Research Center of Medical and Health Big Data, School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, 710048 People’s Republic of China
| |
Collapse
|
112
|
Picard E, Verschoor CP, Ma GW, Pawelec G. Relationships Between Immune Landscapes, Genetic Subtypes and Responses to Immunotherapy in Colorectal Cancer. Front Immunol 2020; 11:369. [PMID: 32210966 PMCID: PMC7068608 DOI: 10.3389/fimmu.2020.00369] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is highly heterogeneous at the genetic and molecular level, which has major repercussions on the efficacy of immunotherapy. A small subset of CRCs exhibit microsatellite instability (MSI), a molecular indicator of defective DNA mismatch repair (MMR), but the majority are microsatellite-stable (MSS). The high tumor mutational burden (TMB) and neoantigen load in MSI tumors favors the infiltration of immune effector cells, and antitumor immune responses within these tumors are strong relative to their MSS counterparts. MSI has emerged as a major predictive marker for the efficacy of immune checkpoint blockade over the last few years and nivolumab or pembrolizumab targeting PD-1 has been approved for patients with MSI refractory or metastatic CRC. However, some MSS tumors show DNA polymerase epsilon (POLE) mutations that also confer a very high TMB and may also be heavily infiltrated by immune cells making them amenable to respond to immune checkpoint inhibitors (ICI). In this review we discuss the role of the different immune landscapes in CRC and their relationships with defined CRC genetic subtypes. We discuss potential reasons why immune checkpoint blockade has met with limited success for the majority of CRC patients, despite the finding that immune cell infiltration of primary non-metastatic tumors is a strong predictive, and prognostic factor for relapse and survival. We then consider in which ways CRC cells develop mechanisms to resist ICI. Finally, we address the latest advances in CRC vaccination and how a personalized neoantigen vaccine strategy might overcome the resistance of MSI and MSS tumors in patients for whom immune checkpoint blockade is not a treatment option.
Collapse
Affiliation(s)
- Emilie Picard
- Health Sciences North Research Institute, Sudbury, ON, Canada
| | | | - Grace W Ma
- Department of Surgery, Health Sciences North, Sudbury, ON, Canada
| | - Graham Pawelec
- Health Sciences North Research Institute, Sudbury, ON, Canada.,Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
113
|
Lee M, Eng G, Barbari SR, Deshpande V, Shcherbakova PV, Gala MK. Homologous Recombination Repair Truncations Predict Hypermutation in Microsatellite Stable Colorectal and Endometrial Tumors. Clin Transl Gastroenterol 2020; 11:e00149. [PMID: 32352724 PMCID: PMC7145036 DOI: 10.14309/ctg.0000000000000149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/13/2020] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Somatic mutations in BRCA1/2 and other homologous recombination repair (HRR) genes have been associated with sensitivity to PARP inhibitors and/or platinum agents in several cancers, whereas hypermutant tumors caused by alterations in POLE or mismatch repair genes have demonstrated robust responses to immunotherapy. We investigated the relationship between somatic truncations in HRR genes and hypermutation in colorectal cancer (CRC) and endometrial cancer (EC). METHODS We analyzed the mutational spectra associated with somatic BRCA1/2 truncations in multiple genomic cohorts (N = 2,335). From these results, we devised a classifier incorporating HRR genes to predict hypermutator status among microsatellite stable (MSS) tumors. Using additional genomic cohorts (N = 1,439) and functional in vivo assays, we tested the classifier to disambiguate POLE variants of unknown significance and identify MSS hypermutators without somatic POLE exonuclease domain mutations. RESULTS Hypermutator phenotypes were prevalent among CRCs with somatic BRCA1/2 truncations (50/62, 80.6%) and ECs with such mutations (44/47, 93.6%). The classifier predicted MSS hypermutators with a cumulative true-positive rate of 100% in CRC and 98.0% in EC and a false-positive rate of 0.07% and 0.63%. Validated by signature analyses of tumor exomes and in vivo assays, the classifier accurately reassigned multiple POLE variants of unknown significance as pathogenic and identified MSS hypermutant samples without POLE exonuclease domain mutations. DISCUSSION Somatic truncations in HRR can accurately fingerprint MSS hypermutators with or without known pathogenic exonuclease domain mutations in POLE and may serve as a low-cost biomarker for immunotherapy decisions in MSS CRC and EC.
Collapse
Affiliation(s)
- Minyi Lee
- Gastrointestinal Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - George Eng
- Gastrointestinal Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephanie R. Barbari
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vikram Deshpande
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Polina V. Shcherbakova
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Manish K. Gala
- Gastrointestinal Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
114
|
Coupez D, Hulo P, Touchefeu Y, Bossard C, Bennouna J. Pembrolizumab for the treatment of colorectal cancer. Expert Opin Biol Ther 2020; 20:219-226. [PMID: 31952453 DOI: 10.1080/14712598.2020.1718095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Colorectal cancer (CRC) is one of the most frequent and lethal cancers in the world, and therapies are still insufficient. Immune checkpoint inhibitors (ICI) in metastatic CRC (mCRC) have not revolutionized treatment to the extent that they have in melanoma or renal carcinoma. Their use is limited to a molecular niche of mCRC with microsatellite instability (MSI). This review summarizes clinical data published with pembrolizumab in mCRC and also tries to identify potential new strategies.Areas covered: This paper focuses on pembrolizumab in mCRC. We screened all trials on PubMed and ClinicalTrials.gov and describe the most significant ones in our opinion.Expert opinion: Pembrolizumab seems to be effective in tumors with MSI-high status. It defines a new horizon for therapeutic strategy called 'agnostic' medicine. For microsatellite stable (MSS) colorectal cancers, the future challenge will be to successfully redraw the immune microenvironment to make it immunogenic with new therapeutic combinations, including ICI.
Collapse
Affiliation(s)
- Dahna Coupez
- Digestive Oncology, Institut Des Maladies De l'Appareil Digestif, Centre Hospitalier Universitaire De Nantes, Nantes, France
| | - Pauline Hulo
- Digestive Oncology, Institut Des Maladies De l'Appareil Digestif, Centre Hospitalier Universitaire De Nantes, Nantes, France.,Centre De Recherche En Cancérologie Et Immunologie Nantes-Angers (CRCINA), INSERM, Université d'Angers, Université De Nantes, Nantes, France
| | - Yann Touchefeu
- Digestive Oncology, Institut Des Maladies De l'Appareil Digestif, Centre Hospitalier Universitaire De Nantes, Nantes, France
| | - Céline Bossard
- Service d'Anatomie Et Cytologie Pathologiques, Centre Hospitalier Universitaire, Nantes, France
| | - Jaafar Bennouna
- Digestive Oncology, Institut Des Maladies De l'Appareil Digestif, Centre Hospitalier Universitaire De Nantes, Nantes, France.,Centre De Recherche En Cancérologie Et Immunologie Nantes-Angers (CRCINA), INSERM, Université d'Angers, Université De Nantes, Nantes, France
| |
Collapse
|
115
|
Huyghe N, Baldin P, Van den Eynde M. Immunotherapy with immune checkpoint inhibitors in colorectal cancer: what is the future beyond deficient mismatch-repair tumours? Gastroenterol Rep (Oxf) 2020; 8:11-24. [PMID: 32104582 PMCID: PMC7034232 DOI: 10.1093/gastro/goz061] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/25/2019] [Accepted: 10/22/2019] [Indexed: 12/17/2022] Open
Abstract
Following initial success in melanoma and lung tumours, immune checkpoint inhibitors (ICIs) are now well recognized as a major immunotherapy treatment modality for multiple types of solid cancers. In colorectal cancer (CRC), the small subset that is mismatch-repair-deficient and microsatellite-instability-high (dMMR/MSI-H) derive benefit from immunotherapy; however, the vast majority of patients with proficient MMR (pMMR) or with microsatellite stable (MSS) CRC do not. Immunoscore and the consensus molecular subtype classifications are promising biomarkers in predicting therapeutic efficacy in selected CRC. In pMRR/MSS CRC, biomarkers are also needed to understand the molecular mechanisms governing immune reactivity and to predict their relationship to treatment. The continuous development of such biomarkers would offer new perspectives and more personalized treatments by targeting oncological options, including ICIs, which modify the tumour-immune microenvironment. In this review, we focus on CRC and discuss the current status of ICIs, the role of biomarkers to predict response to immunotherapy, and the approaches being explored to render pMMR/MSS CRC more immunogenic through the use of combined therapies.
Collapse
Affiliation(s)
- Nicolas Huyghe
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, Brussels, Belgium
| | - Paméla Baldin
- Department of Pathology, Cliniques Universitaires St-Luc, Institut Roi Albert II, Brussels, Belgium
| | - Marc Van den Eynde
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, Brussels, Belgium
- Department of Medical Oncology, Cliniques Universitaires St-Luc, Institut Roi Albert II, Brussels, Belgium
| |
Collapse
|
116
|
Kreidieh M, Mukherji D, Temraz S, Shamseddine A. Expanding the Scope of Immunotherapy in Colorectal Cancer: Current Clinical Approaches and Future Directions. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9037217. [PMID: 32090113 PMCID: PMC7008242 DOI: 10.1155/2020/9037217] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022]
Abstract
The success of immune checkpoint inhibitors (ICIs) in an increasing range of heavily mutated tumor types such as melanoma has culminated in their exploration in different subsets of patients with metastatic colorectal cancer (mCRC). As a result of their dramatic and durable response rates in patients with chemorefractory, mismatch repair-deficient-microsatellite instability-high (dMMR-MSI-H) mCRC, ICIs have become potential alternatives to classical systemic therapies. The anti-programmed death-1 (PD-1) agents, Pembrolizumab and Nivolumab, have been granted FDA approval for this subset of patients. Unfortunately, however, not all CRC cases with the dMMR-MSI-H phenotype respond well to ICIs, and ongoing studies are currently exploring biomarkers that can predict good response to them. Another challenge lies in developing novel treatment strategies for the subset of patients with the mismatch repair-proficient-microsatellite instability-low (pMMR-MSI-L) phenotype that comprises 95% of all mCRC cases in whom treatment with currently approved ICIs has been largely unsuccessful. Approaches aiming at overcoming the resistance of tumors in this subset of patients are being developed including combining different checkpoint inhibitors with either chemotherapy, anti-angiogenic agents, cancer vaccines, adoptive cell transfer (ACT), or bispecific T-cell (BTC) antibodies. This review describes the rationale behind using immunotherapeutics in CRC. It sheds light on the progress made in the use of immunotherapy in the treatment of patients with dMMR-MSI-H CRC. It also discusses emerging approaches and proposes potential strategies for targeting the immune microenvironment in patients with pMMR-MSI-L CRC tumors in an attempt to complement immune checkpoint inhibition.
Collapse
Affiliation(s)
- Malek Kreidieh
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
117
|
Klempner SJ, Fabrizio D, Bane S, Reinhart M, Peoples T, Ali SM, Sokol ES, Frampton G, Schrock AB, Anhorn R, Reddy P. Tumor Mutational Burden as a Predictive Biomarker for Response to Immune Checkpoint Inhibitors: A Review of Current Evidence. Oncologist 2020; 25:e147-e159. [PMID: 31578273 PMCID: PMC6964127 DOI: 10.1634/theoncologist.2019-0244] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Treatment with immune checkpoint inhibitors (ICPIs) extends survival in a proportion of patients across multiple cancers. Tumor mutational burden (TMB)-the number of somatic mutations per DNA megabase (Mb)-has emerged as a proxy for neoantigen burden that is an independent biomarker associated with ICPI outcomes. Based on findings from recent studies, TMB can be reliably estimated using validated algorithms from next-generation sequencing assays that interrogate a sufficiently large subset of the exome as an alternative to whole-exome sequencing. Biological processes contributing to elevated TMB can result from exposure to cigarette smoke and ultraviolet radiation, from deleterious mutations in mismatch repair leading to microsatellite instability, or from mutations in the DNA repair machinery. A variety of clinical studies have shown that patients with higher TMB experience longer survival and greater response rates following treatment with ICPIs compared with those who have lower TMB levels; this includes a prospective randomized clinical trial that found a TMB threshold of ≥10 mutations per Mb to be predictive of longer progression-free survival in patients with non-small cell lung cancer. Multiple trials are underway to validate the predictive values of TMB across cancer types and in patients treated with other immunotherapies. Here we review the rationale, algorithm development methodology, and existing clinical data supporting the use of TMB as a predictive biomarker for treatment with ICPIs. We discuss emerging roles for TMB and its potential future value for stratifying patients according to their likelihood of ICPI treatment response. IMPLICATIONS FOR PRACTICE: Tumor mutational burden (TMB) is a newly established independent predictor of immune checkpoint inhibitor (ICPI) treatment outcome across multiple tumor types. Certain next-generation sequencing-based techniques allow TMB to be reliably estimated from a subset of the exome without the use of whole-exome sequencing, thus facilitating the adoption of TMB assessment in community oncology settings. Analyses of multiple clinical trials across several cancer types have demonstrated that TMB stratifies patients who are receiving ICPIs by response rate and survival. TMB, alongside other genomic biomarkers, may provide complementary information in selecting patients for ICPI-based therapies.
Collapse
Affiliation(s)
- Samuel J. Klempner
- The Angeles Clinic and Research InstituteLos AngelesCaliforniaUSA
- Samuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | | | | | | | - Siraj M. Ali
- Foundation Medicine, Inc.CambridgeMassachusettsUSA
| | | | | | | | | | | |
Collapse
|
118
|
Sveen A, Kopetz S, Lothe RA. Biomarker-guided therapy for colorectal cancer: strength in complexity. Nat Rev Clin Oncol 2020; 17:11-32. [PMID: 31289352 PMCID: PMC7577509 DOI: 10.1038/s41571-019-0241-1] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2019] [Indexed: 12/16/2022]
Abstract
The number of molecularly stratified treatment options available to patients with colorectal cancer (CRC) is increasing, with a parallel rise in the use of biomarkers to guide prognostication and treatment decision-making. The increase in both the number of biomarkers and their use has resulted in a progressively complex situation, evident both from the extensive interactions between biomarkers and from their sometimes complex associations with patient prognosis and treatment benefit. Current and emerging biomarkers also reflect the genomic complexity of CRC, and include a wide range of aberrations such as point mutations, amplifications, fusions and hypermutator phenotypes, in addition to global gene expression subtypes. In this Review, we provide an overview of current and emerging clinically relevant biomarkers and their role in the management of patients with CRC, illustrating the intricacies of biomarker interactions and the growing treatment opportunities created by the availability of comprehensive molecular profiling.
Collapse
Affiliation(s)
- Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research & K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research & K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
119
|
Pang SW, Awi NJ, Armon S, Lim WWD, Low JSH, Peh KB, Peh SC, Teow SY. Current Update of Laboratory Molecular Diagnostics Advancement in Management of Colorectal Cancer (CRC). Diagnostics (Basel) 2019; 10:E9. [PMID: 31877940 PMCID: PMC7168209 DOI: 10.3390/diagnostics10010009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/11/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) continues to be one of the most common cancers globally. The incidence has increased in developing countries in the past few decades, this could be partly attributed to aging populations and unhealthy lifestyles. While the treatment of CRC has seen significant improvement since the advent of target-specific therapies and personalized medicine, CRC is oftentimes detected at late or advanced stages, thereby reducing the efficacy of treatment. Hence, screening for early detection is still the key to combat CRC and to increase overall survival (OS). Considering that the field of medical diagnostics is moving towards molecular diagnostics, CRC can now be effectively screened and diagnosed with high accuracy and sensitivity. Depending on the tumor genotype and genetic profile of the individual, personalized treatments including tyrosine kinase inhibitor therapy and immunotherapy can be administered. Notably, there can be no one single treatment that is effective for all CRC patients due to the variation in tumor genetics, which highlights the importance of molecular diagnostics. This review provides insights on therapeutic modalities, molecular biomarkers, advancement of diagnostic technologies, and current challenges in managing CRC.
Collapse
Affiliation(s)
- Siew-Wai Pang
- Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Jalan Universiti, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Noel Jacques Awi
- Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Jalan Universiti, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Subasri Armon
- Pathology Department, Hospital Kuala Lumpur, Jalan Pahang, Kuala Lumpur 50588, Malaysia
| | - Wendy Wan-Dee Lim
- Sunway Medical Centre, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - John Seng-Hooi Low
- Sunway Medical Centre, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Kaik-Boo Peh
- Mahkota Medical Centre, Mahkota Melaka, Jalan Merdeka, Melaka 75000, Malaysia
| | - Suat-Cheng Peh
- Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Jalan Universiti, Bandar Sunway, Subang Jaya 47500, Malaysia
- Sunway Medical Centre, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Sin-Yeang Teow
- Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Jalan Universiti, Bandar Sunway, Subang Jaya 47500, Malaysia
| |
Collapse
|
120
|
Silberman R, F. Steiner D, Lo AA, Gomez A, Zehnder JL, Chu G, Suarez CJ. Complete and Prolonged Response to Immune Checkpoint Blockade in POLE-Mutated Colorectal Cancer. JCO Precis Oncol 2019; 3:1-5. [DOI: 10.1200/po.18.00214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
121
|
Martin SZ, Wagner DC, Hörner N, Horst D, Lang H, Tagscherer KE, Roth W. Ex vivo tissue slice culture system to measure drug-response rates of hepatic metastatic colorectal cancer. BMC Cancer 2019; 19:1030. [PMID: 31675944 PMCID: PMC6824140 DOI: 10.1186/s12885-019-6270-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/16/2019] [Indexed: 02/08/2023] Open
Abstract
Background The lack of predictive biomarkers or test systems contributes to high failure rates of systemic therapy in metastasized colorectal carcinoma, accounting for a still unfavorable prognosis. Here, we present an ex vivo functional assay to measure drug-response based on a tissue slice culture approach. Methods Tumor tissue slices of hepatic metastases of nine patients suffering from colorectal carcinoma were cultivated for 72 h and treated with different concentrations of the clinically relevant drugs Oxaliplatin, Cetuximab and Pembrolizumab. Easy to use, objective and automated analysis routines based on the Halo platform were developed to measure changes in proliferative activity and the morphometric make-up of the tumor. Apoptotic indices were assessed semiquantitatively. Results Untreated tumor tissue slices showed high morphological comparability with the original “in vivo”-tumor, preserving proliferation and stromal-tumor interactions. All but one patients showed a dosage dependent susceptibility to treatment with Oxaliplatin, whereas only two patients showed responses to Cetuximab and Pembrolizumab, respectively. Furthermore, we identified possible non-responders to Cetuximab therapy in absence of RAS-mutations. Conclusions This is the first time to demonstrate feasibility of the tissue slice culture approach for metastatic tissue of colorectal carcinoma. An automated readout of proliferation and tumor-morphometry allows for quantification of drug susceptibility. This strongly indicates a potential value of this technique as a patient-specific test-system of targeted therapy in metastatic colorectal cancer. Co-clinical trials are needed to customize for clinical application and to define adequate read-out cut-off values.
Collapse
Affiliation(s)
- Steve Z Martin
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany. .,Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Charité Mitte, 10117, Berlin, Germany.
| | - Daniel C Wagner
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Nina Hörner
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Charité Mitte, 10117, Berlin, Germany
| | - Hauke Lang
- Department of General Visceral and Transplantation Surgery, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Katrin E Tagscherer
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
122
|
Yao J, Gong Y, Zhao W, Han Z, Guo S, Liu H, Peng X, Xiao W, Li Y, Dang S, Liu G, Li L, Huang T, Chen S, Song L. Comprehensive analysis of POLE and POLD1 Gene Variations identifies cancer patients potentially benefit from immunotherapy in Chinese population. Sci Rep 2019; 9:15767. [PMID: 31673068 PMCID: PMC6823448 DOI: 10.1038/s41598-019-52414-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/17/2019] [Indexed: 12/31/2022] Open
Abstract
POLE/POLD1 gene variants have been suggested as potential markers for immunotherapy due to their significant association with the tumor mutational burden (TMB), an effective indicator for response prediction in immunotherapy. However, the correlation of POLE/POLD1 variants with MSI, MMR, TMB, MMR-related and key driver gene mutations needs to be defined to support patient recruitment and therapeutic effect assessment in immunotherapy. 1,392 Chinese cancer patients were recruited, and the correlation of POLE/POLD1 variants with existing immunotherapeutic markers and cancer pathways was investigated. A next-generation sequencing panel including 605 cancer-related genes was used for variant sequencing. It was found that the frequency of POLE variants was not statistically different from that in COSMIC database, while the frequency of POLD1 variants was significantly higher in lung cancer. c.857 C > G and c.2091dupC were potential high frequency variants in Chinese cancer patients. Patients carrying POLE damaging variants were significantly younger than POLE/POLD1 WT patients. Patients carrying POLE/POLD1 damaging variants exhibited significantly higher TMB and frequency of MMR gene variants than POLE/POLD1 WT patients. Patients with POLE damaging variants also exhibited significantly higher frequency of driver gene variants than POLE/POLD1 WT patients. Further analysis showed that POLE damaging variants may affect the cancer development through MMR, TGFβ and RTK/RAS/RAF signaling pathways, and POLD1 through MMR pathways. In conclusion, this study identified key characteristics and regions of POLE/POLD1 genes that correlates with TMB, MMR gene mutations and key driver gene mutations, and provided theoretical and practical basis for patient selection based on POLE/POLD1 gene status in immunotherapy.
Collapse
Affiliation(s)
- Jianfei Yao
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China
| | - Yuan Gong
- Department of Gastroenterology, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Wei Zhao
- Department of Thoracic Surgery, Sino-Japanese Friendship Hospital, Jilin University, Changchun, Jilin Province, P.R. China
| | - Zhifeng Han
- Department of Thoracic Surgery, Sino-Japanese Friendship Hospital, Jilin University, Changchun, Jilin Province, P.R. China
| | - Shaohua Guo
- Department of General Surgery, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Hongyi Liu
- Department of General Surgery, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiumei Peng
- Department of Oncology, the Fourth Medical Center of the Chinese PLA General Hospital, Beijing, P.R. China
| | - Wenhua Xiao
- Department of Oncology, the Fourth Medical Center of the Chinese PLA General Hospital, Beijing, P.R. China
| | - Yuemin Li
- Department of Radiotherapy, the Eighth Medical Center of the Chinese PLA General Hospital, Beijing, P.R. China
| | - Shiying Dang
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China
| | - Guifeng Liu
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China
| | - Lifeng Li
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China
| | - Tanxiao Huang
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China
| | - Shifu Chen
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China
| | - Lele Song
- HaploX Biotechnology, Co., Ltd, Shenzhen, P.R. China.
- Department of Radiotherapy, the Eighth Medical Center of the Chinese PLA General Hospital, Beijing, P.R. China.
| |
Collapse
|
123
|
IJsselsteijn ME, Sanz-Pamplona R, Hermitte F, de Miranda NF. Colorectal cancer: A paradigmatic model for cancer immunology and immunotherapy. Mol Aspects Med 2019; 69:123-129. [DOI: 10.1016/j.mam.2019.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/16/2019] [Accepted: 05/24/2019] [Indexed: 12/28/2022]
|
124
|
Guler I, Askan G, Klostergaard J, Sahin IH. Precision medicine for metastatic colorectal cancer: an evolving era. Expert Rev Gastroenterol Hepatol 2019; 13:919-931. [PMID: 31475851 DOI: 10.1080/17474124.2019.1663174] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Metastatic colorectal cancer (CRC) remains a dilemma for cancer researchers with an increasing incidence in the younger patient population. Until the last decade, limited therapeutic options were available for metastatic CRC patients leading to relatively poor clinical outcomes.Areas covered: With advances in genome sequencing technology and reductions in the cost of next-generation sequencing, molecular profiling has become more accessible for cancer researchers and clinical investigators, which has furthered our understanding of the molecular behavior of CRC. This progress has recently translated into significant advances in molecular-based therapeutics and led to the development of new target-specific agents in metastatic CRC patients. In this review article, we extensively elaborate on genomic alterations seen in CRC patients including, but not limited to, EGFR, MMR, BRAF, HER2, NTRKs, FGFR, BRCA1/2, PALB2, POLE, and POLD1 genes, all of which are potentially actionable by either an FDA-approved agent or in a clinical trial setting.Expert opinion: We strongly recommend molecular profiling in metastatic CRC patients during the early course of their disease, as this may provide therapeutic and prognostic information that can guide clinicians to practice precision medicine. Patients with potentially actionable genes should be considered for targeting agents based on molecular alterations.
Collapse
Affiliation(s)
- Irem Guler
- Department of Medicine, Baskent University School of Medicine, Ankara, Turkey
| | - Gokce Askan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jim Klostergaard
- Department of Molecular and Cellular Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ibrahim Halil Sahin
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
125
|
Fakih M, Ouyang C, Wang C, Tu TY, Gozo MC, Cho M, Sy M, Longmate JA, Lee PP. Immune overdrive signature in colorectal tumor subset predicts poor clinical outcome. J Clin Invest 2019; 129:4464-4476. [PMID: 31524634 PMCID: PMC6763253 DOI: 10.1172/jci127046] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
The prognostic value of immune cell infiltration within the tumor microenvironment (TME) has been extensively investigated via histological and genomic approaches. Based on the positive prognostic value of T cell infiltration, Immunoscore has been developed and validated for predicting risk of recurrence for colorectal cancer (CRC). Also, association between a consensus T helper 1 (Th-1) immune response and favorable clinical outcomes has been observed across multiple cancer types. Here, we reanalyzed public genomic data sets from The Cancer Genome Atlas (TCGA) and NCBI Gene Expression Omnibus (NCBI-GEO) and performed multispectral immunohistochemistry (IHC) on a cohort of colorectal tumors. We identified and characterized a risk group, representing approximately 10% of CRC patients, with high intratumoral CD8+ T cell infiltration, but poor prognosis. These tumors included both microsatellite instable (MSI) and stable (MSS) phenotypes and had a high density of tumor-associated macrophages (TAMs) that expressed CD274 (programmed death-ligand 1 [PD-L1]), TGF-β activation, and an immune overdrive signature characterized by the overexpression of immune response and checkpoint genes. Our findings illustrate that CRC patients may have poor prognosis despite high CD8+ T cell infiltration and provide CD274 as a simple biomarker for identifying these patients.
Collapse
Affiliation(s)
- Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, and
| | - Ching Ouyang
- Center for Informatics, City of Hope National Medical Center, Duarte, California, USA
- Department of Computational and Quantitative Medicine
| | - Chongkai Wang
- Department of Medical Oncology and Therapeutics Research, and
| | | | | | - May Cho
- Department of Medical Oncology and Therapeutics Research, and
| | - Marvin Sy
- Department of Medical Oncology and Therapeutics Research, and
| | - Jeffrey A. Longmate
- Department of Computational and Quantitative Medicine
- Division of Biostatistics, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | | |
Collapse
|
126
|
Tintelnot J, Stein A. Immunotherapy in colorectal cancer: Available clinical evidence, challenges and novel approaches. World J Gastroenterol 2019; 25:3920-3928. [PMID: 31413527 PMCID: PMC6689806 DOI: 10.3748/wjg.v25.i29.3920] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/21/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023] Open
Abstract
In contrast to other tumor types, immunotherapy has not yet become a relevant part of the treatment landscape of unselected colorectal cancer. Beside the small subgroup of deficient mismatch repair or microsatellite instable tumors (about 5%) as a surrogate for high mutational burden and subsequently high neoantigen load and immunogenicity, inhibitors of programmed death 1 (PD-1), programmed death ligand 1 (PD-L1) and/or cytotoxic T lymphocyte-associated antigen-4 were not or only modestly effective in metastatic colorectal cancer. Thus, a variety of combination approaches with chemotherapy, targeted therapy, toll-like receptor agonists, local ablation or oncolytic viruses is currently being evaluated in different disease settings. Despite several encouraging single arm data already presented or published, available randomized data are unimpressive. Adding PD-1/PD-L1 inhibitors to fluoropyrimidines and bevacizumab maintenance showed no beneficial impact on delaying progression. In refractory disease, the combination of PD-1/PD-L1 and MEK inhibitor was not different from regorafenib, whereas a PD-1/PD-L1 and cytotoxic T lymphocyte-associated antigen-4 inhibitor combination demonstrated better overall survival compared to supportive care alone. Clinical trials in all disease settings applying different combination approaches are ongoing and may define the role of immunotherapy in colorectal cancer.
Collapse
Affiliation(s)
- Joseph Tintelnot
- Department of Internal Medicine II (Oncology Center), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Alexander Stein
- Department of Internal Medicine II (Oncology Center), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
127
|
Luchini C, Bibeau F, Ligtenberg MJL, Singh N, Nottegar A, Bosse T, Miller R, Riaz N, Douillard JY, Andre F, Scarpa A. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol 2019; 30:1232-1243. [PMID: 31056702 DOI: 10.1093/annonc/mdz116] [Citation(s) in RCA: 657] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancers with a defective DNA mismatch repair (dMMR) system contain thousands of mutations most frequently located in monomorphic microsatellites and are thereby defined as having microsatellite instability (MSI). Therefore, MSI is a marker of dMMR. MSI/dMMR can be identified using immunohistochemistry to detect loss of MMR proteins and/or molecular tests to show microsatellite alterations. Together with tumour mutational burden (TMB) and PD-1/PD-L1 expression, it plays a role as a predictive biomarker for immunotherapy. METHODS To define best practices to implement the detection of dMMR tumours in clinical practice, the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project, based on a systematic review-approach, to generate consensus recommendations on the: (i) definitions related to the concept of MSI/dMMR; (ii) methods of MSI/dMMR testing and (iii) relationships between MSI, TMB and PD-1/PD-L1 expression. RESULTS The MSI-related definitions, for which a consensus frame-work was used to establish definitions, included: 'microsatellites', 'MSI', 'DNA mismatch repair' and 'features of MSI tumour'. This consensus also provides recommendations on MSI testing; immunohistochemistry for the mismatch repair proteins MLH1, MSH2, MSH6 and PMS2 represents the first action to assess MSI/dMMR (consensus with strong agreement); the second method of MSI/dMMR testing is represented by polymerase chain reaction (PCR)-based assessment of microsatellite alterations using five microsatellite markers including at least BAT-25 and BAT-26 (strong agreement). Next-generation sequencing, coupling MSI and TMB analysis, may represent a decisive tool for selecting patients for immunotherapy, for common or rare cancers not belonging to the spectrum of Lynch syndrome (very strong agreement). The relationships between MSI, TMB and PD-1/PD-L1 expression are complex, and differ according to tumour types. CONCLUSIONS This ESMO initiative is a response to the urgent questions raised by the growing success of immunotherapy and provides also important insights on the relationships between MSI, TMB and PD-1/PD-L1.
Collapse
Affiliation(s)
- C Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - F Bibeau
- Department of Pathology, Caen University Hospital, Caen, France
| | - M J L Ligtenberg
- Departments of Human Genetics Radboud university medical center, Nijmegen, The Netherlands; Departments of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - N Singh
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - A Nottegar
- Department of Surgery, San Bortolo Hospital, Vicenza, Italy
| | - T Bosse
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - R Miller
- Department of Oncology, University College London, London, UK
| | - N Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J-Y Douillard
- European Society for Medical Oncology, Lugano, Switzerland
| | - F Andre
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France.
| | - A Scarpa
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
128
|
Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, Miller VA, Lim D, Amanam I, Chao J, Catenacci D, Cho M, Braiteh F, Klempner SJ, Ali SM, Fakih M. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann Oncol 2019; 30:1096-1103. [PMID: 31038663 DOI: 10.1093/annonc/mdz134] [Citation(s) in RCA: 436] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Microsatellite instability (MSI) is a biomarker for response to immune checkpoint inhibitors (ICPIs). PD-1 inhibitors in metastatic colorectal carcinoma (mCRC) with MSI-high (MSI-H) have demonstrated a high disease control rate and favorable progression-free survival (PFS); however, reported response rates to pembrolizumab and nivolumab are variable and often <50%, suggesting that additional predictive biomarkers are needed. METHODS Clinicopathologic data were collected from patients with MSI-H mCRC confirmed by hybrid capture-based next-generation sequencing (NGS) treated with PD-1/L1 inhibitors at five institutes. Tumor mutational burden (TMB) was determined on 0.8-1.1 Mb of sequenced DNA and reported as mutations/Mb. Potential biomarkers of response and time to progression were analyzed by univariate and multivariate analyses. Once TMB was confirmed as a predictive biomarker, a larger dataset of 18 140 unique CRC patients was analyzed to define the relevance of the identified TMB cut-point. RESULTS A total of 22 patients were treated with PD-1/L1 inhibitors including 19 with pembrolizumab monotherapy. Among tested variables, TMB showed the strongest association with objective response (OR; P < 0.001) and PFS, by univariate (P < 0.001) and multivariate analysis (P < 0.01). Using log-rank statistics, the optimal predictive cut-point for TMB was estimated between 37 and 41 mutations/Mb. All 13 TMBhigh cases responded, while 6/9 TMBlow cases had progressive disease. The median PFS for TMBhigh has not been reached (median follow-up >18 months) while the median PFS for TMBlow was 2 months. A TMB of 37.4 mutations/Mb in a large MSI-H mCRC population (821/18, 140 cases; 4.5%) evaluated by NGS corresponded to the 35th percentile cut-point. CONCLUSIONS TMB appears to be an important independent biomarker within MSI-H mCRC to stratify patients for likelihood of response to ICPIs. If validated in prospective studies, TMB may play an important role in guiding the sequencing and/or combinations of ICPIs in MSI-H mCRC.
Collapse
Affiliation(s)
| | - C Ouyang
- Center for Informatics, City of Hope National Medical Center, Duarte; Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte
| | - J Sandhu
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte
| | - E Sokol
- Foundation Medicine, Inc., Cambridge
| | - D Jin
- Foundation Medicine, Inc., Cambridge
| | - J S Ross
- Foundation Medicine, Inc., Cambridge; Department of Pathology, SUNY Upstate Medical University, Syracuse
| | | | - D Lim
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte
| | - I Amanam
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte
| | - J Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte
| | - D Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago
| | - M Cho
- Division of Hematology and Oncology, Department of Internal Medicine, UC Davis Comprehensive Cancer Center, Sacramento
| | - F Braiteh
- Department of Hematology/Oncology, Comprehensive Cancer Centers of Nevada, Las Vegas
| | - S J Klempner
- The Angeles Clinic and Research Institute, Los Angeles, USA
| | - S M Ali
- Foundation Medicine, Inc., Cambridge
| | - M Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte.
| |
Collapse
|
129
|
|
130
|
Ang C, Klempner SJ, Ali SM, Madison R, Ross JS, Severson EA, Fabrizio D, Goodman A, Kurzrock R, Suh J, Millis SZ. Prevalence of established and emerging biomarkers of immune checkpoint inhibitor response in advanced hepatocellular carcinoma. Oncotarget 2019; 10:4018-4025. [PMID: 31258846 PMCID: PMC6592287 DOI: 10.18632/oncotarget.26998] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
The clinical deployment of immune checkpoint inhibitors (ICIs) has created a tandem drive for the identification of biomarkers linked to benefit. Comprehensive genomic profiling was performed to evaluate the frequency of genomic biomarkers of ICI response in 755 patients with advanced hepatocellular carcinoma (HCC). Median age was 62 years’ old, 73% were male, 46% had extrahepatic disease, 107 had documented hepatitis C, 96 had hepatitis B and 4 patients were coinfected. Median tumor mutation burden (TMB) was 4 mutations/Mb and only 6 tumors (0.8%) were TMB-high. Out of 542 cases assessed for microsatellite instability (MSI), one (0.2%) was MSI-high and TMB-high. Twenty-seven (4%) patients had POLE/D alterations. One patient had a pathogenic POLE R762W mutation but TMB was 4 mutations/Mb. Forty percent had DNA damage response gene alterations. In a small case series (N=17) exploring the relationship between biomarkers and ICI response, one patient (TMB 15 mutations/Mb, MSI-low) had a sustained complete response to nivolumab lasting > 2 years. Otherwise there were no significant genomic or TMB differences between responders, progressors, and those with stable disease. Overall, markers of genomic instability were infrequent in this cohort. Larger clinically annotated datasets are needed to explore genomic and non-genomic determinants of ICI response in HCC.
Collapse
Affiliation(s)
- Celina Ang
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Samuel J Klempner
- Department of Medicine, Division of Hematology/Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | | | | | | - Aaron Goodman
- Department of Medicine, Division of Hematology and Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology and Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - James Suh
- Foundation Medicine, Cambridge, MA, USA
| | | |
Collapse
|
131
|
Ciardiello D, Vitiello PP, Cardone C, Martini G, Troiani T, Martinelli E, Ciardiello F. Immunotherapy of colorectal cancer: Challenges for therapeutic efficacy. Cancer Treat Rev 2019; 76:22-32. [DOI: 10.1016/j.ctrv.2019.04.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
132
|
Gegen Qinlian decoction enhances the effect of PD-1 blockade in colorectal cancer with microsatellite stability by remodelling the gut microbiota and the tumour microenvironment. Cell Death Dis 2019; 10:415. [PMID: 31138779 PMCID: PMC6538740 DOI: 10.1038/s41419-019-1638-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
Therapeutic antibodies targeting PD-1 have made major breakthroughs in cancer treatment. However, the majority of colorectal cancer (CRC) cases are microsatellite stable (MSS) and do not respond to anti-PD-1-based immunotherapy. Combination therapy will be an ideal strategy to overcome this limitation. Gegen Qinlian decoction (GQD), a classical traditional Chinese medicine (TCM) formula, has been clinically proven to be effective in the treatment of ulcerative colitis (UC) and type 2 diabetes mellitus. Here, a systemic pharmacological study revealed that GQD acts through multiple targets and pathways in the human body. Combination therapy with GQD and anti-mouse PD-1 potently inhibited the growth of CT26 tumours in a xenograft model. Gut microbiota analysis revealed that combination therapy with GQD and anti-mouse PD-1 significantly enriched for s__Bacteroides_acidifaciens and s__uncultured_organism_g__norank_f__Bacteroidales_S24-7_group. Based on metabolomic analyses, profoundly altered metabolites were identified in the combination therapy group. Two metabolic signalling pathways, namely, glycerophospholipid metabolism and sphingolipid metabolism, were explored. In particular, we found that combination therapy with GQD and anti-mouse PD-1 significantly increased the proportion of CD8+ T cells in peripheral blood and tumour tissues. Direct treatment with GQD and anti-mouse PD-1 increased the expression of IFN-γ, which is a critical factor in antitumour immunotherapy. In addition, combination therapy with GQD and anti-mouse PD-1 downregulated PD-1 and increased IL-2 levels, suggesting that the combination therapy could effectively restore T-cell functions by suppressing inhibitory checkpoints. The application of the Chinese medicinal formula GQD with PD-1 blockade-based immunotherapy can be a novel therapeutic strategy for CRC patients with MSS tumours.
Collapse
|
133
|
Ma K, Jin Q, Wang M, Li X, Zhang Y. Research progress and clinical application of predictive biomarker for immune checkpoint inhibitors. Expert Rev Mol Diagn 2019; 19:517-529. [PMID: 31079502 DOI: 10.1080/14737159.2019.1617702] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have emerged as epochal milestones in the field of anti-cancer immunotherapy. With promising clinical effectiveness, ICIs can significantly prolong the overall survival of patients with advanced cancer of different types. Although their remarkable effectiveness has been demonstrated in clinical application, ICIs display limitations in terms of unique response patterns. Only a subset of patients exhibits objective responses, while others show rapid disease progression. Considering that there is a fair representation of both subsets of patients (responders and non-responders), clinicians ought to effectively stratify patients who will potentially benefit from ICI therapy, and optimize a strategy for patient selection. Areas covered: In this review, the authors have summarized several key factors involved in the biomarker development of ICI therapy, such as neoantigen production and presentation, the tumor microenvironment, and alternation in specific gene signaling pathways. Expert opinion: Considering the extreme complexity of the immune system, a single biomarker may fail to appropriately stratify patients for ICI therapy. Therefore, future biomarker research should focus on designing an integrated biomarker system that will successfully guide combination therapies to overcome resistance to immunotherapy.
Collapse
Affiliation(s)
- Ke Ma
- a Department of Pharmacology , School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University , Shenyang , China
| | - Qingqing Jin
- a Department of Pharmacology , School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University , Shenyang , China
| | - Miao Wang
- a Department of Pharmacology , School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University , Shenyang , China
| | - Xin Li
- a Department of Pharmacology , School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University , Shenyang , China
| | - Yuyang Zhang
- a Department of Pharmacology , School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University , Shenyang , China
| |
Collapse
|
134
|
Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front Oncol 2019; 9:396. [PMID: 31139574 PMCID: PMC6527887 DOI: 10.3389/fonc.2019.00396] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
There are a wide range of therapies for metastatic colorectal cancer (CRC) available, but outcomes remain suboptimal. Learning the role of the immune system in cancer development and progression led to advances in the treatment over the last decade. While the field is rapidly evolving, PD-1, and PD-L1 inhibitors have a leading role amongst immunomodulatory agents. They act against pathways involved in adaptive immune suppression resulting in immune checkpoint blockade. Immunotherapy has been slow to impact the management of this patient population due to disappointing results, mainly when used broadly. Nevertheless, some patients with microsatellite-instability-high (MSI-H) or mismatch repair-deficient (dMMR) CRC appear to be susceptible to checkpoint inhibitors with objective and sustained clinical responses, providing a new therapeutic option for patients with advanced disease. This article provides a comprehensive review of the early and late phase trials with the updated data of PD-1/PD-L1 inhibitors alone or in combination with other therapies (immunotherapy, targeted therapy and chemotherapy). While data is still limited, many ongoing trials are underway, testing the efficacy of these agents in CRC. Current and future challenges of PD-1 and PD-L1 inhibitors are also discussed.
Collapse
Affiliation(s)
- André F. Oliveira
- Serviço de Oncologia Médica, Centro Hospitalar Universitário Algarve, Faro, Portugal
| | | | | |
Collapse
|
135
|
Wang Y, Johnson DB, Lu S, Diaz LA, Xu Y, Balko JM. Tumor genomic alterations in severe-combined immunodeficiency bare-lymphocyte syndrome genes are associated with high mutational burden and disproportional neo-antigen rates. J Immunother Cancer 2019; 7:123. [PMID: 31064401 PMCID: PMC6503546 DOI: 10.1186/s40425-019-0584-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
The progression of cancer requires mutational adaptation to permit unrestrained proliferation. A fraction of cancer mutations are oncogenic drivers, while others are putative 'passengers' that do not contribute to oncogenesis. However, altered peptides arising from passenger mutations may bind MHCs and activate non-self immunologic signals (i.e. neoantigens), thus requiring immunoediting for cancer persistence. Disruption of antigen processing machinery in tumor cells may diminish this requirement. Here, we show that rare mutations in antigen processing machinery are associated with high mutational burden and increased predicted neoantigen load, providing insights into the mechanisms of high mutation burden in some patients.
Collapse
Affiliation(s)
- Yu Wang
- 0000 0004 1936 9916grid.412807.8Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232 USA ,0000 0004 1936 9916grid.412807.8Center for Quantitative Sciences, Vanderbilt University Medical Center, 2200 Pierce Ave, 777 PRB, Nashville, TN 37232–6307 USA
| | - Douglas B. Johnson
- 0000 0004 1936 9916grid.412807.8Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Steve Lu
- 0000 0001 2171 9311grid.21107.35Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Luis A. Diaz
- 0000 0001 2171 9952grid.51462.34Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Center for Quantitative Sciences, Vanderbilt University Medical Center, 2200 Pierce Ave, 777 PRB, Nashville, TN, 37232-6307, USA.
| | - Justin M. Balko
- 0000 0004 1936 9916grid.412807.8Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 USA ,0000 0004 1936 9916grid.412807.8Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| |
Collapse
|
136
|
Lasabová Z, Kalman M, Holubeková V, Grendár M, Kašubová I, Jašek K, Meršaková S, Malicherová B, Baranenko D, Adamek M, Kruzliak P, Plank L. Mutation analysis of POLE gene in patients with early-onset colorectal cancer revealed a rare silent variant within the endonuclease domain with potential effect on splicing. Clin Exp Med 2019; 19:393-400. [PMID: 31049795 DOI: 10.1007/s10238-019-00558-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022]
Abstract
The colorectal cancer harbor germline, somatic or epimutations in mismatch repair genes, MUTYH or POLE gene, which lead to the hypermutated and ultramutator phenotypes with increased immune response. The mutations in POLE gene were reported to occur more frequently in early-onset colorectal cancer (EOCRC), and the patients are strong candidates for checkpoint inhibitor therapy. Here, we report mutation analysis within the endonuclease domain of the POLE gene in the cohort of patients with EOCRC in order to identify recurrent or new mutations and evaluate their association with the presence of tumor-infiltrating lymphocytes (TILs) and peritumoral lymphoid reaction. We have shown a significant association between MSI tumors and TILs (p = 0.004). Using sensitive single-tube nested PCR with subsequent Sanger sequencing, we have found in one female patient diagnosed at age 48 with rectal adenocarcinoma with mucinous elements staged pT3pN2pM1 a silent variant within the exon 9 NM_006231.3 c.849 C > T, NP_00622.2 p.Leu283 = recorded in dSNP as rs1232888774 with MAF = 0.00002. In silico prediction, result showed possible involvement into splicing; therefore, this rare variant can be involved into EOCRC pathogenesis. In the time of precise medicine, it is important to develop screening strategies also for less common conditions such as EOCRC allowing to predict tailored therapy for younger patients suffering from CRC that harbor mutations in the POLE gene.
Collapse
Affiliation(s)
- Zora Lasabová
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Maláhora 4C, 03601, Martin, Slovakia. .,Department of Molecular Biology and Genomics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia.
| | - Michal Kalman
- Department of Pathological Anatomy, Jessenius Faculty of Medicine University Hospital in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Veronika Holubeková
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Maláhora 4C, 03601, Martin, Slovakia
| | - Marián Grendár
- Department of Bioinformatics, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Ivana Kašubová
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Maláhora 4C, 03601, Martin, Slovakia
| | - Karin Jašek
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Maláhora 4C, 03601, Martin, Slovakia
| | - Sandra Meršaková
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Maláhora 4C, 03601, Martin, Slovakia
| | - Bibiana Malicherová
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Maláhora 4C, 03601, Martin, Slovakia
| | - Denis Baranenko
- International Research Centre "Biotechnologies of the Third Millennium", ITMO University, Saint-Petersburg, Russian Federation
| | - Mariusz Adamek
- Department of Thoracic Surgery, Faculty of Medicine and Dentistry, Medical University of Silesia, Katowice, Poland
| | - Peter Kruzliak
- Department of Internal Medicine, Brothers of Mercy Hospital, Brno, Czech Republic.
| | - Lukáš Plank
- Department of Pathological Anatomy, Jessenius Faculty of Medicine University Hospital in Martin, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
137
|
Lu L, Huang H, Zhou J, Ma W, Mackay S, Wang Z. BRCA1 mRNA expression modifies the effect of T cell activation score on patient survival in breast cancer. BMC Cancer 2019; 19:387. [PMID: 31023256 PMCID: PMC6482542 DOI: 10.1186/s12885-019-5595-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/09/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Effector CD8+ T cell activation and its cytotoxic function to eradicate tumor cells depend on the T cell recognition of tumor neoantigens, and are positively associated with improved survival in breast cancer. Tumor suppressor BRCA1 and cell cycle regulator CCND1 play a critical role in maintaining genome integrity and tumorigenesis, respectively. However, it is still unclear how BRCA1 and CCND1 expression levels affect the effect of T cell activation on breast cancer patient survival. METHODS The interactions between T cell activation status and either BRCA1 or CCND1 expression were evaluated using Kaplan-Meier survival curves and multivariate Cox regression models in a public dataset with 1088 breast cancer patients. RESULTS Among the patients with low BRCA1 or CCND1 expression, the Activation group showed better overall survival than the Exhaustion group. Adjusted hazards ratios were 0.43 (95% CI: 0.20-0.93) in patients with a low BRCA1 level, and 0.39 (95% CI: 0.19-0.81) in patients with a low CCND1 level, respectively. There was a significant trend in both subgroups (p-trend = 0.011 in the low BRCA1 group, and p-trend = 0.009 in the low CCND1 group). In contrast, there is no significant association in patients with either high BRCA1 or high CCND1 levels. There is a significant interaction between T cell activation status and BRCA1 level (p = 0.009), but not between T cell activation status and CCND1 level (p = 0.135). CONCLUSIONS BRCA1 expression modified the effect of T cell activation status on patient survival in breast cancer, suggesting that the existence of neoantigens and the enhancement of neoantigen presentation in combination with immune checkpoint blockade may have synergistic effects on patient outcome.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Center for Biomedical Data Science, Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520-8034 USA
| | - Huatian Huang
- Guizhou Qianxinan People’s Hospital, Xingyi, 652400 Guizhou China
| | - Jing Zhou
- Isoplexis Corporation, 35 NE Industrial Road, Branford, CT 06405 USA
| | - Wenxue Ma
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093 USA
| | - Sean Mackay
- Isoplexis Corporation, 35 NE Industrial Road, Branford, CT 06405 USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, School of Medicine, Center for Biomedical Data Science, Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520-8034 USA
| |
Collapse
|
138
|
Sandhu J, Lavingia V, Fakih M. Systemic treatment for metastatic colorectal cancer in the era of precision medicine. J Surg Oncol 2019; 119:564-582. [PMID: 30802315 DOI: 10.1002/jso.25421] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/10/2019] [Indexed: 01/05/2023]
Abstract
The treatment of metastatic colorectal cancer has evolved over the last two decades with the FDA approval of several cytotoxic, biological, and targeted agents. In this paper, we review the impact of sidedness, RAS, BRAF, HER-2, and other immune biomarkers on metastatic colorectal cancer treatment selection and sequencing in both the palliative and curative intent settings.
Collapse
Affiliation(s)
- Jaideep Sandhu
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | | | - Marwan Fakih
- Medical Oncology and Therapeutics Research, Briskin Center for Clinical Research, GI Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
139
|
Gbolahan O, O’Neil B. Update on systemic therapy for colorectal cancer: biologics take sides. Transl Gastroenterol Hepatol 2019; 4:9. [PMID: 30976712 PMCID: PMC6414333 DOI: 10.21037/tgh.2019.01.12] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 12/13/2022] Open
Abstract
Over the last decade, progress in the management of metastatic colorectal cancer (CRC) has focused on the development of biologic therapy in addition to the back bone of combination chemotherapy. Anti-epidermal growth factor receptor (EGFR) antibodies and agents targeting angiogenesis are widely used in the clinic, and more recently, in a subset of patients with mismatch repair (MMR) deficient cancer, immunotherapy with immune check point inhibitors have been integrated into clinical practice. The major challenge with the use of these biologic therapies is determining predictive biomarkers to optimize patient selection. In this review, we discuss the most recent updates in the use of biologic therapy in CRC. We review data on the role of primary tumor location (PTL) (sidedness) as predictive biomarker and recent advances in treatment of CRC with BRAF mutation.
Collapse
Affiliation(s)
- Olumide Gbolahan
- Department of Hematology and Oncology, University of Alabama, Birmingham School of Medicine, Birmingham, AL, USA
| | - Bert O’Neil
- Indiana University School of Medicine, Indiana Cancer Pavilion, Indianapolis, IN, USA
| |
Collapse
|
140
|
Martin B, Märkl B. Immunologic Biomarkers and Biomarkers for Immunotherapies in Gastrointestinal Cancer. Visc Med 2019; 35:3-10. [PMID: 31312644 DOI: 10.1159/000496565] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal (GI) cancers contribute significantly to the worldwide cancer burden. Pathologic evaluation is indispensable for the estimation of prognosis and therapeutic strategy. At present, immunotherapies are evolving into efficient therapeutic approaches, which are accompanied by the need for biomarkers to predict therapy response. In colorectal cancers, the only predictive biomarker for Food and Drug Administration-approved immunotherapy is the mismatch repair status. Besides, pathogenic polymerase epsilon mutations, tumor mutational burden, neoantigen load, and features of the immune contexture could soon find their way into clinical routine. Furthermore, in colorectal cancer, the Immunoscore, which is defined by the amount of CD3+ and CD8+ T-cells in the tumor center as well as at the infiltrative margin, might supplement the TNM system in the future (as TNM-Immune). This immunologic biomarker was shown to be impressively prognostic and predictive in colorectal cancer. In conclusion, there is increasing evidence of immunologic as well as predictive biomarkers for immunotherapies in GI cancers. Nevertheless, future progress is necessary for the variety of current advances to be implemented in clinical care.
Collapse
Affiliation(s)
- Benedikt Martin
- Institute of Pathology, University Clinic Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Institute of Pathology, University Clinic Augsburg, Augsburg, Germany
| |
Collapse
|
141
|
Cohen R, Heran M, Pudlarz T, Hilmi M, Tournigand C, André T, Rousseau B. Traitement des cancers colorectaux par immunothérapie : aller au-delà de MSI avec la classification moléculaire (CMS) et la charge mutationnelle. Bull Cancer 2019; 106:151-161. [DOI: 10.1016/j.bulcan.2018.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 01/27/2023]
|
142
|
Lee EK, Lindeman NI, Matulonis UA, Konstantinopoulos PA. POLE-mutated clear cell cervical cancer associated with in-utero diethylstilbestrol exposure. Gynecol Oncol Rep 2019; 28:15-17. [PMID: 30733993 PMCID: PMC6357694 DOI: 10.1016/j.gore.2019.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/24/2022] Open
Abstract
We report an extraordinary case of a woman, exposed to diethylstilbestrol in utero, who developed clear cell adenocarcinoma of the cervix with a concurrent polymerase-Ɛ (POLE) somatic mutation. The tumor exhibited the classic phenotypic characteristics of POLE-mutated tumors originating from other organs (e.g. the uterus or the colon) including increased tumor infiltrating lymphocytes and high PD-L1 expression and has remained in remission since completion of primary therapy for >4 years. This case highlights the importance of next generation sequencing in unraveling the biology of rare tumors and supports that the presence of a POLE mutation and the associated ultramutated state confers a unique phenotype of higher immunogenicity and possibly improved prognosis in a tissue-agnostic manner, i.e. regardless of the type of cancer where the POLE mutation is present. We report a POLE-mutated clear cell cervical cancer associated with in-utero DES. Tumor exhibited increased tumor infiltrating lymphocytes and PD-L1 expression. Patient remains in remission for ≥4 years after standard therapy. POLE mutation confers a phenotype of higher immunogenicity and improved outcome.
Collapse
Affiliation(s)
- Elizabeth K Lee
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
143
|
Patel JN, Fong MK, Jagosky M. Colorectal Cancer Biomarkers in the Era of Personalized Medicine. J Pers Med 2019; 9:E3. [PMID: 30646508 PMCID: PMC6463111 DOI: 10.3390/jpm9010003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The 5-year survival probability for patients with metastatic colorectal cancer has not drastically changed over the last several years, nor has the backbone chemotherapy in first-line disease. Nevertheless, newer targeted therapies and immunotherapies have been approved primarily in the refractory setting, which appears to benefit a small proportion of patients. Until recently, rat sarcoma (RAS) mutations remained the only genomic biomarker to assist with therapy selection in metastatic colorectal cancer. Next generation sequencing has unveiled many more potentially powerful predictive genomic markers of therapy response. Importantly, there are also clinical and physiologic predictive or prognostic biomarkers, such as tumor sidedness. Variations in germline pharmacogenomic biomarkers have demonstrated usefulness in determining response or risk of toxicity, which can be critical in defining dose intensity. This review outlines such biomarkers and summarizes their clinical implications on the treatment of colorectal cancer. It is critical that clinicians understand which biomarkers are clinically validated for use in practice and how to act on such test results.
Collapse
Affiliation(s)
- Jai N Patel
- Department of Cancer Pharmacology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA.
| | - Mei Ka Fong
- Department of Pharmacy, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA.
| | - Megan Jagosky
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA.
| |
Collapse
|
144
|
Liu S, Gӧnen M, Stadler ZK, Weiser MR, Hechtman JF, Vakiani E, Wang T, Vyas M, Joneja U, Al-Bayati M, Segal NH, Smith JJ, King S, Guercio S, Ntiamoah P, Markowitz AJ, Zhang L, Cercek A, Garcia-Aguilar J, Saltz LB, Diaz LA, Klimstra DS, Shia J. Cellular localization of PD-L1 expression in mismatch-repair-deficient and proficient colorectal carcinomas. Mod Pathol 2019; 32:110-121. [PMID: 30166615 PMCID: PMC6309293 DOI: 10.1038/s41379-018-0114-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Blockade of the interaction between PD-1 and its ligands PD-L1 has shown clinical efficacy across several tumor types, especially in mismatch-repair-deficient colorectal carcinoma. The aim of this study was to examine the pattern and cellular localization of PD-L1 expression in the different molecular subtypes of mismatch-repair-deficient colorectal cancers vs. their mismatch-repair-proficient counterparts. PD-L1/SATB2 double-antibody-immunohistochemistry was utilized to distinguish tumor cell from immune cell staining. We observed in our series of 129 colorectal adenocarcinomas that PD-L1 expression occurred primarily in tumor-associated-immune cells and most prominently at the tumor-stroma-interface of the invasive front. The level of invasive front immune cell staining was significantly higher in mismatch-repair-deficient tumors compared to mismatch-repair-proficient tumors (p < 0.001), but no difference was observed among the different subtypes of mismatch-repair-deficient tumors: Lynch syndrome-associated vs. MLH1-methylated vs. unexplained. While selected mismatch-repair-proficient tumors exhibited unusually high tumor-infiltrating-lymphocytes and had high level immune cell PD-L1 expression, a positive correlation between PD-L1 expression and high lymphocyte count was detected only in mismatch-repair-deficient tumors (r = 0.39, p < 0.001) and not in mismatch-repair-proficient tumors. Notably, true tumor cell PD-L1 expression in colorectal carcinoma was rare, present in only 3 of 129 tumors (2.3%): 2 MLH1-methylated and 1 mismatch-repair-proficient with high tumor-infiltrating-lymphocytes; and the staining in the tumor cells in all 3 was diffuse (>=50% of the tumor). These findings may serve to inform further efforts aiming to evaluate PD-L1 immunohistochemistry vis-à-vis molecular sub-classification as predictive biomarkers in the treatment of colorectal carcinoma.
Collapse
Affiliation(s)
- Sandy Liu
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Mithat Gӧnen
- Department of Biostatistics, Memorial Sloan Kettering
Cancer Center, New York, NY
| | - Zsofia K. Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Martin R. Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Jaclyn F. Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Tao Wang
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Monika Vyas
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Upasana Joneja
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Moataz Al-Bayati
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Neil H. Segal
- Department of Medicine, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - J. Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Sarah King
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Shanna Guercio
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Peter Ntiamoah
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Arnold J. Markowitz
- Department of Medicine, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Liying Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer
Center, New York, NY
| | | | - Leonard B. Saltz
- Department of Medicine, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Luis A. Diaz
- Department of Medicine, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - David S. Klimstra
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
145
|
Abstract
Genomic information is increasingly being incorporated into clinical cancer care. Large-scale sequencing efforts have deepened our understanding of the genomic landscape of cancer and contributed to the expanding catalog of alterations being leveraged to aid in cancer diagnosis, prognosis, and treatment. Genomic profiling can provide clinically relevant information regarding somatic point mutations, copy number alterations, translocations, and gene fusions. Genomic features, such as mutational burden, can also be measured by more comprehensive sequencing strategies and have shown value in informing potential treatment options. Ongoing clinical trials are evaluating the use of molecularly targeted agents in genomically defined subsets of cancers within and across tumor histologies. Continued advancements in clinical genomics promise to further expand the application of genomics-enabled medicine to a broader spectrum of oncology patients.
Collapse
Affiliation(s)
- Alison Roos
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Sara A Byron
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
146
|
Xiong D, Wang Y, Singavi AK, Mackinnon AC, George B, You M. Immunogenomic Landscape Contributes to Hyperprogressive Disease after Anti-PD-1 Immunotherapy for Cancer. iScience 2018; 9:258-277. [PMID: 30439581 PMCID: PMC6234258 DOI: 10.1016/j.isci.2018.10.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/14/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022] Open
Abstract
Although PD-1-blocking immunotherapies demonstrate significant therapeutic promise, a subset of the patients could develop hyperprogressive disease (HPD) with accelerated tumor growth after anti-PD1 immunotherapy. To elucidate the underlying mechanisms, we compared the mutational and transcriptional landscapes between the pre- and post-therapy tumors of two patients developing HPD after anti-PD-1 immunotherapy. In post-therapy HPD tumors, somatic mutations were found in known cancer genes, including tumor suppressor genes such as TSC2 and VHL, along with transcriptional upregulation of oncogenic pathways, including IGF-1, ERK/MAPK, PI3K/AKT, and TGF-β. We found that post-therapy HPD tumors were less immunogenic than pre-therapy tumors, concurrent with an increased presence of ILC3 cells, a subset of innate lymphoid cells. We also developed a gene expression signature predictive of HPD. In summary, we identified the genomics and immune features associated with HPD, which may help identify patients at risk of adverse clinical outcome after anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Donghai Xiong
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yian Wang
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arun K Singavi
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alexander C Mackinnon
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ben George
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ming You
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
147
|
Dienstmann R, Salazar R, Tabernero J. Molecular Subtypes and the Evolution of Treatment Decisions in Metastatic Colorectal Cancer. Am Soc Clin Oncol Educ Book 2018; 38:231-238. [PMID: 30231342 DOI: 10.1200/edbk_200929] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Colorectal cancer (CRC) has clinically relevant molecular heterogeneity at multiple levels: genomics, epigenomics, transcriptomics, and microenvironment features. Genomic events acquired during carcinogenesis remain drivers of cancer progression in the metastatic setting. For example, KRAS and NRAS mutations define a population refractory to epidermal growth factor receptor monoclonal antibodies, BRAFV600E mutations associate with poor outcomes under standard therapies and response to targeted inhibitors in combinations, and HER2 amplifications confer unique sensitivity to double HER2 blockade. Multiple rare gene alterations driving resistance to epidermal growth factor receptor monoclonal antibodies have been described, with substantial overlap in primary and acquired mechanisms, in line with a clonal selection process. In this context, sequential analysis of circulating tumor DNA has the potential to guide drug development in a treatment-refractory setting. Rare kinase fusion events and complex alterations in genes involved in DNA damage repair have been described, with emerging evidence for targetability. On the other hand, transcriptomic subtypes and pathway activation signatures have also shown prognostic and potential predictive value in metastatic CRC. These markers reflect stromal and immune microenvironment interactions with cancer cells. For example, the microsatellite instable or POLE ultramutant CRC population is particularly sensitive to immune checkpoint inhibitors, whereas tumors with a mesenchymal phenotype are characterized by activation of immunosuppressive molecules that mandate stratified development of novel immunotherapy combinations. Here, we review the expanding landscape of targetable oncogenic alterations and signatures in metastatic CRC and discuss the clinical implementation of novel molecular diagnostic tests.
Collapse
Affiliation(s)
- Rodrigo Dienstmann
- From the Oncology Data Science Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain; Sage Bionetworks, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Medical Oncology, Catalan Institute of Oncology, Oncobell Program, L'Hospitalet de Llobregat, CIBERONC, Barcelona, Spain; Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Ramon Salazar
- From the Oncology Data Science Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain; Sage Bionetworks, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Medical Oncology, Catalan Institute of Oncology, Oncobell Program, L'Hospitalet de Llobregat, CIBERONC, Barcelona, Spain; Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Josep Tabernero
- From the Oncology Data Science Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain; Sage Bionetworks, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Medical Oncology, Catalan Institute of Oncology, Oncobell Program, L'Hospitalet de Llobregat, CIBERONC, Barcelona, Spain; Medical Oncology Department, Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| |
Collapse
|
148
|
Goel G. Molecular characterization and biomarker identification in colorectal cancer: Toward realization of the precision medicine dream. Cancer Manag Res 2018; 10:5895-5908. [PMID: 30510457 PMCID: PMC6250110 DOI: 10.2147/cmar.s162967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is a major public health problem, both in the USA and globally. Over the past 20 years, significant advances have been made in the treatment of patients with metastatic CRC (mCRC). Recent efforts in the field of biomarkers have focused on the development of molecular diagnostics to define the subset of patients with mCRC that is likely to derive most benefit from anti-EGFR therapy. Herein, we review the recent advancements in molecular stratification of CRC and the role of current as well as emerging biomarkers in this disease. It is now clear that the presence of activating mutations in the KRAS and NRAS genes serves as reliable predictive markers for resistance to anti-EGFR therapy in mCRC. It is also clear that further improvements in the survival of mCRC patients will probably be made possible only with identification of new predictive molecular biomarkers and their evaluation using rational and innovative clinical trials. The recent advances in DNA sequencing technology and "omics"-based approaches have provided promising new strategies for the development of novel molecular biomarkers in this disease.
Collapse
Affiliation(s)
- Gaurav Goel
- Division of Hematology-Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,
| |
Collapse
|
149
|
Kondou R, Iizuka A, Nonomura C, Miyata H, Ashizawa T, Nagashima T, Ohshima K, Urakami K, Kusuhara M, Yamaguchi K, Akiyama Y. Classification of tumor microenvironment immune types based on immune response-associated gene expression. Int J Oncol 2018; 54:219-228. [PMID: 30387832 DOI: 10.3892/ijo.2018.4617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/27/2018] [Indexed: 11/06/2022] Open
Abstract
In 2014, the Shizuoka Cancer Center launched project High‑tech Omics‑based Patient Evaluation (HOPE), which features whole exome sequencing (WES) and gene expression profiling (GEP) of fresh surgical specimens from cancer patients. With the development of clinical trials of programmed death‑1 (PD‑1)/PD‑ligand 1 (PD‑L1) blockade, PD‑L1 expression and a high tumor mutation burden become possible biomarkers that could be used to predict immune responses. In this study, based on WES and GEP data from 1,734 tumors from the HOPE project, we established a tumor microenvironment (TME) immune‑type classification consisting of 4 types to evaluate the immunological status of cancer patients and analyze immunological pathways specific for immune types. Project HOPE was conducted in accordance with the Ethical Guidelines for Human Genome and Genetic Analysis Research with the approval of the Institutional Review Board. Based on the expression level of the PD‑L1 and CD8B genes, the immunological status was divided into 4 types as follows: A, PD‑L1+CD8B+; B, PD‑L1+CD8B‑; C, PD‑L1‑CD8B‑; and D, PD‑L1‑CD8B+. Type A, with PD‑L1+ and CD8B+, exhibited an upregulation of cytotoxic T lymphocyte (CTL) killing‑associated genes, T‑cell activation genes, antigen‑presentation and dendritic cell (DC) maturation genes, and T‑cell‑attracting chemokine genes, which promoted Th1 antitumor responses. By contrast, type C, with PD‑L1‑ and CD8B‑, exhibited a low expression of T‑cell‑activating genes and an upregulation of cancer driver gene signaling, which suggested an immune‑suppressive status. With regard to hypermutator tumors, PD‑L1+ hypermutator cases exhibited a specific upregulation of the IL6 gene compared with the PD‑L1‑ cases. On the whole, our data indicate that the classification of the TME immune types may prove to be a useful tool for evaluating the immunological status and predicting antitumor responses and prognosis.
Collapse
Affiliation(s)
- Ryota Kondou
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Akira Iizuka
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Chizu Nonomura
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | | | - Keiichi Ohshima
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Kenichi Urakami
- Cancer Diagnostic Research Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Masatoshi Kusuhara
- Regional Resources Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Office of the President, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| |
Collapse
|
150
|
Lee L, Ali S, Genega E, Reed D, Sokol E, Mathew P. Aggressive-Variant Microsatellite-Stable POLE Mutant Prostate Cancer With High Mutation Burden and Durable Response to Immune Checkpoint Inhibitor Therapy. JCO Precis Oncol 2018; 2:1-8. [DOI: 10.1200/po.17.00097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Lisa Lee
- Lisa Lee, Elizabeth Genega, Dallas Reed, and Paul Mathew, Tufts Medical Center, Boston; Siraj Ali and Ethan Sokol, Foundation Medicine, Cambridge, MA
| | - Siraj Ali
- Lisa Lee, Elizabeth Genega, Dallas Reed, and Paul Mathew, Tufts Medical Center, Boston; Siraj Ali and Ethan Sokol, Foundation Medicine, Cambridge, MA
| | - Elizabeth Genega
- Lisa Lee, Elizabeth Genega, Dallas Reed, and Paul Mathew, Tufts Medical Center, Boston; Siraj Ali and Ethan Sokol, Foundation Medicine, Cambridge, MA
| | - Dallas Reed
- Lisa Lee, Elizabeth Genega, Dallas Reed, and Paul Mathew, Tufts Medical Center, Boston; Siraj Ali and Ethan Sokol, Foundation Medicine, Cambridge, MA
| | - Ethan Sokol
- Lisa Lee, Elizabeth Genega, Dallas Reed, and Paul Mathew, Tufts Medical Center, Boston; Siraj Ali and Ethan Sokol, Foundation Medicine, Cambridge, MA
| | - Paul Mathew
- Lisa Lee, Elizabeth Genega, Dallas Reed, and Paul Mathew, Tufts Medical Center, Boston; Siraj Ali and Ethan Sokol, Foundation Medicine, Cambridge, MA
| |
Collapse
|