1
|
Younis A, Gribben J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. IMMUNO 2024; 4:186-210. [DOI: 10.3390/immuno4030013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) are a promising form of immunotherapy that have significantly changed the therapeutic landscape for many advanced cancers. They have shown unique clinical benefit against a broad range of tumour types and a strong overall impact on survival in studied patient populations. However, there are still many limitations holding back this immunotherapy from reaching its full potential as a possible curative option for advanced cancer patients. A great deal of research is being undertaken in the hope of driving advancements in this area, building a better understanding of the mechanisms behind immune checkpoint inhibition and ultimately developing more effective, safer, and wider-reaching agents. Taking into account the current literature on this topic, this review aims to explore in depth the basis of the use of ICIs in the treatment of advanced cancers, evaluate its efficacy and safety, consider its current limitations, and finally reflect on what the future holds for this very promising form of cancer immunotherapy.
Collapse
Affiliation(s)
- Abdullah Younis
- Barts and the London School of Medicine and Dentistry, London E1 2AD, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
| |
Collapse
|
2
|
Ho V, Chung L, Wilkinson K, Ma Y, Rutland T, Lea V, Lim SH, Abubakar A, Ng W, Lee M, Roberts TL, Becker TM, Mackenzie S, Chua W, Lee CS. Microsatellite Instability Testing and Prognostic Implications in Colorectal Cancer. Cancers (Basel) 2024; 16:2005. [PMID: 38893125 PMCID: PMC11171323 DOI: 10.3390/cancers16112005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Given the crucial predictive implications of microsatellite instability (MSI) in colorectal cancer (CRC), MSI screening is commonly performed in those with and at risk for CRC. Here, we compared results from immunohistochemistry (IHC) and the droplet digital PCR (ddPCR) MSI assay on formalin-fixed paraffin-embedded tumor samples from 48 patients who underwent surgery for colon and rectal cancer by calculating Cohen's kappa measurement (k), revealing high agreement between the methods (k = 0.915). We performed Kaplan-Meier survival analyses and univariate and multivariate Cox regression to assess the prognostic significance of ddPCR-based MSI and to identify clinicopathological features associated with CRC outcome. Patients with MSI-high had better overall survival (OS; p = 0.038) and disease-free survival (DFS; p = 0.049) than those with microsatellite stability (MSS). When stratified by primary tumor location, right-sided CRC patients with MSI-high showed improved DFS, relative to those with MSS (p < 0.001), but left-sided CRC patients did not. In multivariate analyses, MSI-high was associated with improved OS (hazard ratio (HR) = 0.221, 95% confidence interval (CI): 0.026-0.870, p = 0.042), whereas the loss of DNA mismatch repair protein MutL homolog 1 (MLH1) expression was associated with worse OS (HR = 0.133, 95% CI: 0.001-1.152, p = 0.049). Our results suggest ddPCR is a promising tool for MSI detection. Given the opposing effects of MSI-high and MLH1 loss on OS, both ddPCR and IHC may be complementary for the prognostic assessment of CRC.
Collapse
Affiliation(s)
- Vincent Ho
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
| | - Liping Chung
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
| | - Kate Wilkinson
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Yafeng Ma
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Tristan Rutland
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Vivienne Lea
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Stephanie H. Lim
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, NSW 2560, Australia
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Askar Abubakar
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
| | - Weng Ng
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Mark Lee
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Tara L. Roberts
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Therese M. Becker
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Scott Mackenzie
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia (Y.M.); (T.R.); (V.L.); (A.A.); (T.L.R.); (T.M.B.); (S.M.); (W.C.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (K.W.); (S.H.L.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| |
Collapse
|
3
|
Wang Y, Yang S, Wan L, Ling W, Chen H, Wang J. New developments in the mechanism and application of immune checkpoint inhibitors in cancer therapy (Review). Int J Oncol 2023; 63:86. [PMID: 37326100 PMCID: PMC10308343 DOI: 10.3892/ijo.2023.5534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has been demonstrated in the treatment of numerous types of cancer and ICIs have remained a key focus of cancer research. However, improvements in survival rates only occur in a subset of patients, due to the complexity of drug resistance. Therefore, further investigations are required to identify predictive biomarkers that distinguish responders and non‑responders. Combined therapeutics involving ICIs and other modalities demonstrate potential in overcoming resistance to ICIs; however, further preclinical and clinical trials are required. Concurrently, prompt recognition and intervention of immune‑related adverse events are crucial to optimize the use of ICIs in clinical treatment. The present study aimed to review the current literature surrounding the mechanisms and application of ICIs, with the aim of providing a theoretical basis for clinicians.
Collapse
Affiliation(s)
- Yanjun Wang
- Department of Urology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510062
| | - Shuo Yang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036
| | - Li Wan
- Department of Endocrinology and Metabolism, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Wei Ling
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Jinghua Wang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080
| |
Collapse
|
4
|
Ruff SM, Pawlik TM. A Review of Translational Research for Targeted Therapy for Metastatic Colorectal Cancer. Cancers (Basel) 2023; 15:cancers15051395. [PMID: 36900187 PMCID: PMC10000872 DOI: 10.3390/cancers15051395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Colorectal cancer is the third most common cause of cancer-related death in the United States, with 20% of patients presenting with metastatic disease at the time of diagnosis. Metastatic colon cancer is often treated with a combination of surgery, systemic therapy (chemotherapy, biologic therapy, immunotherapy), and/or regional therapy (hepatic artery infusion pumps). Utilizing the molecular and pathologic features of the primary tumor to tailor treatment for patients may improve overall survival. Rather than a "one size fits all" approach, a more nuanced treatment plan guided by the unique features of a patient's tumor and the tumor's microenvironment can more effectively treat the disease. Basic science work to elucidate new drug targets, understand mechanisms of evasion, and develop drugs and drug combinations is critical to inform clinical trials and identify novel, effective therapies for metastatic colorectal cancer. Through the lens of key targets for metastatic colorectal cancer, this review discusses how work in the basic science lab translates into clinical trials.
Collapse
|
5
|
Maiorano BA, Parisi A, Maiello E, Ciardiello D. The Interplay between Anti-Angiogenics and Immunotherapy in Colorectal Cancer. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101552. [PMID: 36294987 PMCID: PMC9604892 DOI: 10.3390/life12101552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Colorectal cancer is a frequent and lethal neoplasm. The tumor often creates new vessels to grow and spread—a process called ‘angiogenesis’. Therefore, drugs blocking angiogenesis are effective against this malignancy. On the other side, immune checkpoint inhibitors, which unleash the immune system to fight against tumors, have limited efficacy in patients carrying instability of DNA regions called microsatellites. However, there is an interaction between angiogenic factors and the immune system. This gives a chance to combine anti-angiogenic agents and immune checkpoint inhibitors to improve the efficacy of treating this malignancy. Abstract Angiogenesis, a hallmark of cancer, plays a fundamental role in colorectal cancer (CRC). Anti-angiogenic drugs and chemotherapy represent a standard of care for treating metastatic disease. Immune checkpoint inhibitors (ICIs) have changed the therapeutic algorithm of many solid tumors. However, the efficacy of ICIs is limited to mCRC patients carrying microsatellite instability (MSI-H), which represent approximately 3–5% of mCRC. Emerging evidence suggests that anti-angiogenic drugs could exhibit immunomodulatory properties. Thus, there is a strong rationale for combining anti-angiogenics and ICIs to improve efficacy in the metastatic setting. Our review summarizes the pre-clinical and clinical evidence regarding the combination of anti-angiogenics and ICIs in mCRC to deepen the possible application in daily clinical practice.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Correspondence:
| | - Alessandro Parisi
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, 60126 Ancona, Italy
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Evaristo Maiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Davide Ciardiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Medical Oncology Unit, Department of Precision Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| |
Collapse
|
6
|
Yuan J, Li J, Gao C, Jiang C, Xiang Z, Wu J. Immunotherapies catering to the unmet medical need of cold colorectal cancer. Front Immunol 2022; 13:1022190. [PMID: 36275766 PMCID: PMC9579278 DOI: 10.3389/fimmu.2022.1022190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
As a common malignant tumor of gastrointestinal tract, the incidence of colorectal cancer (CRC) has gradually increased in recent years. In western developed countries, it has even become the second largest malignant tumor next to lung cancer. Immunotherapy is a hot topic in the field of cancer therapy, including immune checkpoint blockade (ICB), adoptive cell therapy (ACT), cancer vaccines and cytokines, aiming to improve the ability of the immune system to recognize, target and eliminate cancer cells. However, cold CRC, which accounts for a high proportion of CRC, is not so reactive to it. The development of immunotherapy to prevent cancer cells from forming “immune escape” pathways to the immune system in cold CRC, has been under increasing study attention. There is proof that an organic combination of radiotherapy, chemotherapy, and several immunotherapies can considerably boost the immune system’s capacity to eradicate tumor cells. In this review, we summarized the role of immunotherapy in colorectal cancer. In addition, we propose a breakthrough and strategy to improve the role of immunotherapy in cold CRC based on its characteristics.
Collapse
Affiliation(s)
- Jun Yuan
- Department of Clinical Laboratory, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, China
| | - Jiarui Li
- Zhejiang University School of Medicine, Hangzhou, China
| | - Ce Gao
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Chun Jiang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Jian Wu, ; Ze Xiang,
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
- *Correspondence: Jian Wu, ; Ze Xiang,
| |
Collapse
|
7
|
Microsatellite Instability: From the Implementation of the Detection to a Prognostic and Predictive Role in Cancers. Int J Mol Sci 2022; 23:ijms23158726. [PMID: 35955855 PMCID: PMC9369169 DOI: 10.3390/ijms23158726] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 02/07/2023] Open
Abstract
Microsatellite instability (MSI) has been identified in several tumors arising from either germline or somatic aberration. The presence of MSI in cancer predicts the sensitivity to immune checkpoint inhibitors (ICIs), particularly PD1/PD-L1 inhibitors. To date, the predictive role of MSI is currently used in the selection of colorectal cancer patients for immunotherapy; moreover, the expansion of clinical trials into other cancer types may elucidate the predictive value of MSI for non-colorectal tumors. In clinical practice, several assays are used for MSI testing, including immunohistochemistry (IHC), polymerase chain reaction (PCR) and next-generation sequencing (NGS). In this review, we provide an overview of MSI in various cancer types, highlighting its potential predictive/prognostic role and the clinical trials performed. Finally, we focus on the comparison data between the different assays used to detect MSI in clinical practice.
Collapse
|
8
|
Waidhauser J, Nerlinger P, Sommer F, Wolf S, Eser S, Löhr P, Rank A, Märkl B. Circulating Lymphocytes Reflect the Local Immune Response in Patients with Colorectal Carcinoma. Diagnostics (Basel) 2022; 12:diagnostics12061408. [PMID: 35741218 PMCID: PMC9221878 DOI: 10.3390/diagnostics12061408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 12/04/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) correlate with the number and size of the surrounding lymph nodes in patients with colorectal carcinoma (CRC) and reflect the quality of the antitumor immune response. In this prospective study, we analyzed whether this response correlated with the circulating lymphocytes in peripheral blood (PB). In 47 patients with newly diagnosed CRC, flow cytometry was performed to analyze the B cells, T cells, NK cells, and a variety of their subsets in PB. The results were correlated with TILs in the resected tumor and with the number and size of the surrounding lymph nodes in nodal negative (N- patients (LN5: number of lymph nodes measuring ≥5 mm) and the metastasis-to-lymph node size ratio (MSR) in nodal positive patients (N+). Differences between the number of TILs could be seen between N+ and N- patients, dependent on the LN5 and MSR categories, with higher values in N- cases and in patients with a higher LN5 category or a lower MSR. Additionally, higher values of various circulating lymphocyte subgroups were observed in these patients. For the total PB lymphocytes, CD8 cells, and some of their subgroups, a positive correlation with the TILs was found. This study shows that circulating lymphocytes—in particular, cytotoxic T cells—correlate with the local antitumor immune response displayed by TILs and lymph node activation. Our findings indicate that local and generalized antitumor immune responses are concordant with their different components.
Collapse
Affiliation(s)
- Johanna Waidhauser
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
- Correspondence:
| | - Pia Nerlinger
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
| | - Florian Sommer
- Department of General, Visceral and Transplant Surgery, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (F.S.); (S.W.)
| | - Sebastian Wolf
- Department of General, Visceral and Transplant Surgery, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (F.S.); (S.W.)
| | - Stefan Eser
- Department of Gastroenterology and Infectious Diseases, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany;
| | - Phillip Löhr
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
| | - Andreas Rank
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany;
| |
Collapse
|
9
|
Lungulescu C, Croitoru VM, Volovat SR, Cazacu IM, Turcu-Stiolica A, Gheonea DI, Sur D, Lungulescu CV. An Insight into Deficient Mismatch Repair Colorectal Cancer Screening in a Romanian Population-A Bi-Institutional Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:847. [PMID: 34441055 PMCID: PMC8401048 DOI: 10.3390/medicina57080847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022]
Abstract
Background and Objectives: Colorectal cancer (CRC) can be classified as mismatch-repair-deficient (dMMR) with high levels of microsatellite instability (MSI-H), or mismatch-repair-proficient (pMMR) and microsatellite stable (MSS). Approximately 15% of patients have microsatellite instability (MSI). MSI-H tumors are associated with a high mutation burden. Monoclonal antibodies that block immune checkpoints can induce long-term durable responses in some patients. Pembrolizumab is the first checkpoint inhibitor approved in the EU to treat dMMR-MSI-H metastatic CRC. Materials and Methods: Our study assesses the regional variability of MSI-H colorectal cancer tumors in Romania. Formalin-fixed, paraffin-embedded (FFPE) tissue blocks containing tumor samples from 90 patients diagnosed with colorectal cancer were collected from two tertiary referral Oncology Centers from Romania. Tissues were examined for the expression loss of MMR proteins (MLH1, PMS2, MSH2, MSH6) using immunohistochemistry or MSI status using polymerase chain reaction (PCR), respectively. Results: MSI-H was detected in 19 (21.1%) patients. MSI-H was located more in ascending colon (36.8% vs. 9.9%, p-value = 0.0039) and less in sigmoid (5.3% vs. 33.8%, p-value = 0.0136) than MSS patients. Most patients were stage II for MSI-H (42.1%) as well as for MSS (56.3%), with significant more G1 (40.9% vs. 15.8%, p-value = 0.0427) for MSS patients. Gender, N stage, and M stage were identified as significant prognostic factors in multivariate analysis. MSI status was not a statistically significant predictor neither in univariate analysis nor multivariate analysis. Conclusion: Considering the efficacy of PD-1 inhibitor in metastatic CRC with MSI-H or dMMR, and its recent approval in EU, it is increasingly important to understand the prevalence across tumor stage, histology, and demographics, since our study displayed higher regional MSI-H prevalence (21%) compared to the literature.
Collapse
Affiliation(s)
- Cristina Lungulescu
- Doctoral School, University of Medicine and Pharmacy Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Vlad Mihai Croitoru
- Department of Oncology, Fundeni Clinical Institute, 258 Fundeni Str., 022238 Bucharest, Romania; (V.M.C.); (I.M.C.)
| | - Simona Ruxandra Volovat
- Department of Medical Oncology, University of Medicine and Pharmacy Grigore T Popa Iasi, 700115 Iasi, Romania;
| | - Irina Mihaela Cazacu
- Department of Oncology, Fundeni Clinical Institute, 258 Fundeni Str., 022238 Bucharest, Romania; (V.M.C.); (I.M.C.)
| | - Adina Turcu-Stiolica
- Department of Pharmacoeconomics, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Dan Ionut Gheonea
- Department of Gastroenterology, University of Medicine and Pharmacy Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Daniel Sur
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania
| | - Cristian Virgil Lungulescu
- Department of Oncology, University of Medicine and Pharmacy Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| |
Collapse
|
10
|
Chaput L, Jordheim LP. [Current landscape of biomarker development for immune checkpoint inhibitors targeting PD-1/PD-L1 pathway in oncology]. Therapie 2021; 76:597-615. [PMID: 34332787 DOI: 10.1016/j.therap.2021.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/25/2021] [Accepted: 06/29/2021] [Indexed: 12/26/2022]
Abstract
The immune checkpoints inhibitors targeting PD-1 or PD-L1 represent a new paradigm in the cancer treatment strategy. However, some populations of patients do not benefit from these agents. The identification of predictive biomarkers appears as an essential step for the treatment pathway, to guarantee the access to an evidence-based medicine accounting for the potential toxicity profile, the cost for the healthcare system and the clinical benefit eventually provided by these new drugs. In this review, we propose, based on scientific literature and industrial communications, an overview of the current landscape of predictive biomarkers related to PD-1 or PD-L1 inhibitors efficacy, validated or under development, their evidence level, and limits accounting for identified or potential confounding factors. Our paper shows that, despite the important amount of work performed in this field, there is not yet a validated and efficient solution for the prediction of the activity and/or the toxicity of anti-PD-1 and anti-PD-L1 antibodies.
Collapse
Affiliation(s)
- Lisa Chaput
- Université Lyon, université Claude-Bernard Lyon 1, faculté de pharmacie de Lyon, ISPB, 69008 Lyon, France
| | - Lars Petter Jordheim
- Université Lyon, université Claude-Bernard Lyon 1, faculté de pharmacie de Lyon, ISPB, 69008 Lyon, France; Université Lyon, université Claude-Bernard Lyon 1, INSERM 1052, CNRS 5286, centre Léon-Bérard, centre de recherche en cancérologie de Lyon, 69008 Lyon, France.
| |
Collapse
|
11
|
Bindal P, Gray JE, Boyle TA, Florou V, Puri S. Biomarkers of therapeutic response with immune checkpoint inhibitors. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1040. [PMID: 34277840 PMCID: PMC8267267 DOI: 10.21037/atm-20-6396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICPIs) have revolutionized the treatment paradigm of a wide range of malignancies with durable responses seen in even advanced, refractory cancers. Unfortunately, only a small proportion of patients with cancer derive meaningful benefit to ICPI therapy, and its use is also limited by significant immune and financial toxicities. Thus, there is a critical need for the development of biomarkers to reliably predict response to ICPI therapy. Only a few biomarkers are validated and approved for use with currently Food and Drug administration (FDA)-approved ICPIs. The development and broad application of biomarkers is limited by the lack of complete understanding of the complex interactions of tumor-host environment, the effect of immunotherapies on these already complex interactions, a lack of standardization and interpretation of biomarker assays across tumor types. Despite these challenges, the field of identifying predictive biomarkers is evolving at an unprecedented pace leaving the clinician responsible for identifying the patients that may derive optimal benefit from ICPIs. In this review, we provide clinicians with a current and practical update on the key, clinically relevant biomarkers of response to ICPIs. We categorize the current and emerging biomarkers of response to ICPIs in four major categories that govern anticancer response—the inflamed tumor, tumor antigens, immune suppression, and overall host environment.
Collapse
Affiliation(s)
- Poorva Bindal
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Vaia Florou
- Division of Medical Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | - Sonam Puri
- Division of Medical Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
12
|
Lin SY, Hang JF, Lin YY, Lai CR, Ho HL, Chou TY. Diffuse Intratumoral Stromal Inflammation in Ovarian Clear Cell Carcinoma is Associated With Loss of Mismatch Repair Protein and High PD-L1 Expression. Int J Gynecol Pathol 2021; 40:148-155. [PMID: 32897958 DOI: 10.1097/pgp.0000000000000682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ovarian clear cell carcinoma (OCCC) is an aggressive chemotherapy-resistant cancer with limited treatment options, and some OCCCs have mismatch repair (MMR) deficiency (MMRD). Emerging evidence has revealed that various cancers with MMRD are susceptible to anti-programmed death-1/programmed death ligand-1 (anti-PD-1/PD-L1) immunotherapy, and certain histologic features are associated with MMRD. However, few studies have addressed this in OCCC. We reviewed 76 OCCCs for tumor-associated inflammation (intratumoral stromal inflammation and peritumoral lymphocytes) and performed immunohistochemistry for 4 MMR proteins and PD-L1. MMR-deficient OCCCs were analyzed for microsatellite instability (MSI), and those with MLH1 loss were tested for MLH1 promoter methylation. No patients fulfilled the Amsterdam II criteria for the diagnosis of Lynch syndrome. Four (5.3%) tumors showed diffuse intratumoral stromal inflammation obliterating the tumor-stroma interfaces, and none had peritumoral lymphoid aggregates. MMRD was found in 2 (2.6%) tumors; one had MLH1/PMS2 loss (MSI-high and MLH1 promoter methylation was detected) and the other had MSH2/MSH6 loss (MSI-low). Twenty (26.3%) tumors showed tumoral PD-L1 expression ≥1%. Both MMR-deficient tumors showed diffuse intratumoral stromal inflammation and tumoral PD-L1 expression ≥50%. Three of the 4 (75%) tumors with diffuse intratumoral stromal inflammation also showed tumoral PD-L1 expression ≥50%. None of the tumors without diffuse intratumoral stromal inflammation showed MMRD (P=0.021) or tumoral PD-L1 expression ≥50% (P=0.0001). We identified a strong correlation among diffuse intratumoral stromal inflammation, MMRD, and high tumoral PD-L1 expression in a small but significant subset of OCCCs. Histologic evaluation can facilitate patient selection for subsequent anti-PD-1/PD-L1 immunotherapy.
Collapse
|
13
|
The Role of Creatine in the Development and Activation of Immune Responses. Nutrients 2021; 13:nu13030751. [PMID: 33652752 PMCID: PMC7996722 DOI: 10.3390/nu13030751] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/06/2023] Open
Abstract
The use of dietary supplements has become increasingly common over the past 20 years. Whereas supplements were formerly used mainly by elite athletes, age and fitness status no longer dictates who uses these substances. Indeed, many nutritional supplements are recommended by health care professionals to their patients. Creatine (CR) is a widely used dietary supplement that has been well-studied for its effects on performance and health. CR also aids in recovery from strenuous bouts of exercise by reducing inflammation. Although CR is considered to be very safe in recommended doses, a caveat is that a preponderance of the studies have focused upon young athletic individuals; thus there is limited knowledge regarding the effects of CR on children or the elderly. In this review, we examine the potential of CR to impact the host outside of the musculoskeletal system, specifically, the immune system, and discuss the available data demonstrating that CR can impact both innate and adaptive immune responses, together with how the effects on the immune system might be exploited to enhance human health.
Collapse
|
14
|
Sadeghi Rad H, Bazaz SR, Monkman J, Ebrahimi Warkiani M, Rezaei N, O'Byrne K, Kulasinghe A. The evolving landscape of predictive biomarkers in immuno-oncology with a focus on spatial technologies. Clin Transl Immunology 2020; 9:e1215. [PMID: 33251010 PMCID: PMC7680923 DOI: 10.1002/cti2.1215] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/16/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapies have shown long-lasting and unparalleled responses for cancer patients compared to conventional therapy. However, they seem to only be effective in a subset of patients. Therefore, it has become evident that a greater understanding of the tumor microenvironment (TME) is required to understand the nuances which may be at play for a favorable outcome to therapy. The immune contexture of the TME is an important factor in dictating how well a tumor may respond to immune checkpoint inhibitors. While traditional immunohistochemistry techniques allow for the profiling of cells in the tumor, this is often lost when tumors are analysed using bulk tissue genomic approaches. Moreover, the actual cellular proportions, cellular heterogeneity and deeper spatial distribution are lacking in characterisation. Advances in tissue interrogation technologies have given rise to spatially resolved characterisation of the TME. This review aims to provide an overview of the current methodologies that are used to profile the TME, which may provide insights into the immunopathology associated with a favorable outcome to immunotherapy.
Collapse
Affiliation(s)
| | - Sajad Razavi Bazaz
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
| | - James Monkman
- The School of Biomedical Science, Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Translational Research InstituteWoolloongabbaQLDAustralia
| | - Majid Ebrahimi Warkiani
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
- Institute of Molecular MedicineSechenov UniversityMoscowRussia
| | - Nima Rezaei
- School of MedicineTehran University of Medical SciencesTehranIran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
- Research Center for ImmunodeficienciesChildren's Medical CenterTehran University of Medical SciencesTehranIran
| | - Ken O'Byrne
- The School of Biomedical Science, Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Translational Research InstituteWoolloongabbaQLDAustralia
- Princess Alexandra HospitalWoolloongabbaQLDAustralia
| | - Arutha Kulasinghe
- The School of Biomedical Science, Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Translational Research InstituteWoolloongabbaQLDAustralia
- Institute for Molecular BiosciencesUniversity of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
15
|
Baliu-Piqué M, Pandiella A, Ocana A. Breast Cancer Heterogeneity and Response to Novel Therapeutics. Cancers (Basel) 2020; 12:E3271. [PMID: 33167363 PMCID: PMC7694303 DOI: 10.3390/cancers12113271] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Targeted cancer therapies against oncogenic drivers are actively being developed and tested in clinical trials. Targeting an oncogenic driver may only prove effective if the mutation is present in most tumoral cells. Therefore, highly heterogeneous tumors may be refractory to these therapies. This makes tumor heterogeneity a major challenge in cancer therapy. Although heterogeneity has traditionally been attributed to genetic diversity within cancer cell populations, it is now widely recognized that human cancers are heterogeneous in almost all distinguishable phenotypic characteristics. Understanding the genetic variability and also the non-genetic influences of tumor heterogeneity will provide novel insights into how to reverse therapeutic resistance and improve cancer therapy.
Collapse
Affiliation(s)
- Mariona Baliu-Piqué
- Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos and CIBERONC, 28040 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer and CIBERONC, CSIC-IBSAL, 37007 Salamanca, Spain;
| | - Alberto Ocana
- Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos and CIBERONC, 28040 Madrid, Spain;
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomedicas, Castilla-La Mancha University (CRIB-UCLM), 02008 Albacete, Spain
| |
Collapse
|
16
|
Vainer ED, Kania-Almog J, Zatara G, Levin Y, Vainer GW. Novel Proteome Extraction Method Illustrates a Conserved Immunological Signature of MSI-H Colorectal Tumors. Mol Cell Proteomics 2020; 19:1619-1631. [PMID: 32641473 PMCID: PMC8015011 DOI: 10.1074/mcp.ra120.002152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/29/2020] [Indexed: 11/14/2022] Open
Abstract
Using a simple, environment friendly proteome extraction (TOP), we were able to optimize the analysis of clinical samples. Using our TOP method we analyzed a clinical cohort of microsatellite stable (MSS) and unstable (MSI-H) colorectal carcinoma (CRC). We identified a tumor cell specific, STAT1-centered, immune signature expressed by the MSI-H tumor cells. We then showed that long, but not short, exposure to Interferon-γ induces a similar signature in vitro We identified 10 different temporal protein expression patterns, classifying the Interferon-γ protein temporal regulation in CRC. Our data sheds light on the changes that tumor cells undergo under long-term immunological pressure in vivo, the importance of STAT proteins in specific biological scenarios. The data generated could help find novel clinical biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Elez D Vainer
- Department of Gastroenterology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Juliane Kania-Almog
- Tel Aviv Sourasky Medical Center, Sackler School of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ghadeer Zatara
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yishai Levin
- De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Gilad W Vainer
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
17
|
Biomarkers for immune checkpoint therapy targeting programmed death 1 and programmed death ligand 1. Biomed Pharmacother 2020; 130:110621. [PMID: 34321165 DOI: 10.1016/j.biopha.2020.110621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Rapidly increasing usages of immune checkpoint therapy for cancer treatment, particularly monoclonal antibodies that target programmed cell death-1 (PD-1) and its ligand PD-L1, have been achieved due to startling durable therapeutic efficacy with limited toxicity. The therapeutics significantly prolonged the overall survival and progression free survival of patients across multiple cancer types. However, the objective response rate of patients receiving this kind of treatment is substantially low. Therefore, it is of great importance to exploit reliable biomarkers that can robustly predict the therapeutic effects. Several biomarkers have been characterized for the selection of patients, which is mainly based on immunological and genetic criteria. Herein, we focus on the current progress regarding the biomarkers for anti-PD-1/PD-L1 therapy.
Collapse
|
18
|
The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis. Sci Rep 2020; 10:3360. [PMID: 32099066 PMCID: PMC7042281 DOI: 10.1038/s41598-020-60255-4] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 02/03/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are an important histopathologic feature of colorectal cancer that confer prognostic information. Previous clinical and epidemiologic studies have found that the presence and quantification of tumor-infiltrating lymphocytes are significantly associated with disease-specific and overall survival in colorectal cancer. We performed a systematic review and meta-analysis, establishing pooled estimates for survival outcomes based on the presence of TILs in colon cancer. PubMed (Medline), Embase, Cochrane Library, Web of Science, and ClinicalTrials.gov were searched from inception to April 2017. Studies were included, in which the prognostic significance of intratumoral tumor infiltrating lymphocytes, as well as subsets of CD3, CD8, FOXP3, CD45R0 lymphocytes, were determined within the solid tumor center, the invasive margin, and tumor stroma. Random-effects models were calculated to estimated summary effects using hazard ratios. Forty-three relevant studies describing 21,015 patients were included in our meta-analysis. The results demonstrate that high levels of generalized TILS as compared to low levels had an improved overall survival (OS) with a HR of 0.65 (p = <0.01). In addition, histologically localized CD3+ T-cells at the tumor center were significantly associated with better disease-free survival (HR = 0.46, 95% CI 0.36–0.61, p = 0.05), and CD3 + cells at the invasive margin were associated with improved disease-free survival (HR = 0.57, 95% CI 0.38–0.86, p = 0.05). CD8+ T-cells at the tumor center had statistically significant prognostic value on cancer-specific survival and overall survival with HRs of 0.65 (p = 0.02) and 0.71 (p < 0.01), respectively. Lastly, FOXP3+ T-cells at the tumor center were associated with improved prognosis for cancer-specific survival (HR = 0.65, p < 0.01) and overall survival (HR = 0.70, p < 0.01). These findings suggest that TILs and specific TIL subsets serve as prognostic biomarkers for colorectal cancer.
Collapse
|
19
|
Otoshi T, Nagano T, Tachihara M, Nishimura Y. Possible Biomarkers for Cancer Immunotherapy. Cancers (Basel) 2019; 11:cancers11070935. [PMID: 31277279 PMCID: PMC6678720 DOI: 10.3390/cancers11070935] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/18/2019] [Accepted: 06/30/2019] [Indexed: 01/05/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have drastically changed the clinical care of cancer. Although cancer immunotherapy has shown promise in various types of malignancies, thus far, the proportion of patients who can benefit from ICIs is relatively small. Immune-related adverse events and high cost are unavoidable problems. Therefore, biomarkers defining patients that are most likely to benefit from ICIs are urgently needed. The expression of programmed cell death-ligand 1 (PD-L1) is a logical biomarker for the prediction of response to anti-PD1/PD-L1 immunotherapies. However, its usefulness is currently debatable because of its varied definition, threshold, and spatial/temporal heterogeneity. Recently, it was reported that the tumor mutational burden, expression of neoantigens, mismatch repair status, and specific gene mutations may be markers for the success of treatment with ICIs. Moreover, it was suggested that the fecal microbiota prior to immunotherapy may play an important role in predicting the efficacy of ICIs. In this review, we focused on these potential biomarkers for cancer immunotherapy reported in recent clinical articles. Further studies are warranted to develop a predictive model using these biomarkers, with the aim of practicing precision medicine in cancer immunotherapy.
Collapse
Affiliation(s)
- Takehiro Otoshi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
20
|
Climent M, Ryan ÉJ, Stakelum Á, Khaw YL, Creavin B, Lloyd A, Alhassan D, Mohan HM, Kennelly R, Sheahan K, Winter DC. Systemic inflammatory response predicts oncological outcomes in patients undergoing elective surgery for mismatch repair-deficient colorectal cancer. Int J Colorectal Dis 2019; 34:1069-1078. [PMID: 30993458 DOI: 10.1007/s00384-019-03274-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION A variety of inflammatory scoring systems and their prognostic value have been reported in many solid organ cancers. This study aimed to examine the association between the systemic and local inflammatory responses, and oncological outcomes in patients undergoing elective surgery for mismatch repair-deficient (dMMR) phenotype colorectal cancer (CRC). MATERIALS AND METHODS Consecutive patients undergoing resection for dMMR CRC were identified from a prospectively maintained database and compared with a cohort of patients with proficient mismatch repair system tumours. Systemic inflammatory response was assessed by the modified Glasgow prognostic score (mGPS), neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio, lymphocyte-monocyte ratio, C-reactive protein/albumin ratio, prognostic index and prognostic nutritional index. Local inflammatory response was defined by the presence of tumour infiltrating lymphocytes, tumour infiltrating neutrophils, plasma cells or macrophages at the invasive front. The inflammatory infiltrate was assessed using the Klintrup-Mäkinen (KM) score. RESULTS On univariable analysis, preoperative NLR ≥ 5 (hazard ratio [HR] 2.5; 95% confidence interval [CI] 1.25-5.19; p = 0.007) and mGPS (HR 1.6; 95% CI 1.1-2.6; p = 0.03) predicted worse overall survival, but only NLR was associated with greater recurrence (HR 3.6; 95% CI 1.5-8.8; p = 0.004). Increased local inflammatory response, as measured by KM score (HR 0.31; 95% CI 0.1-0.7; p = 0.009) and the presence of macrophages in the peritumoral infiltrate (HR 0.17; 95% CI 0.07-0.3; p < 0.001), was associated with better outcomes. NLR was the only independent prognostic factor of overall and disease-free survival. CONCLUSION Systemic inflammatory response predicts oncological outcomes in CRC patients, but only NLR has prognostic value in the dMMR group.
Collapse
Affiliation(s)
- Marta Climent
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.
| | - Éanna J Ryan
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Áine Stakelum
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Yi Ling Khaw
- Department of Histopathology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Ben Creavin
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Angus Lloyd
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dalal Alhassan
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Helen M Mohan
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Rory Kennelly
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kieran Sheahan
- Department of Histopathology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Des C Winter
- Centre for Colorectal Disease, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
21
|
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
|
22
|
Abstract
Checkpoint inhibitor-based immunotherapies that target cytotoxic T lymphocyte antigen 4 (CTLA4) or the programmed cell death 1 (PD1) pathway have achieved impressive success in the treatment of different cancer types. Yet, only a subset of patients derive clinical benefit. It is thus critical to understand the determinants driving response, resistance and adverse effects. In this Review, we discuss recent work demonstrating that immune checkpoint inhibitor efficacy is affected by a combination of factors involving tumour genomics, host germline genetics, PD1 ligand 1 (PDL1) levels and other features of the tumour microenvironment, as well as the gut microbiome. We focus on recently identified molecular and cellular determinants of response. A better understanding of how these variables cooperate to affect tumour-host interactions is needed to optimize the implementation of precision immunotherapy.
Collapse
Affiliation(s)
- Jonathan J Havel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diego Chowell
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
23
|
De' Angelis GL, Bottarelli L, Azzoni C, De' Angelis N, Leandro G, Di Mario F, Gaiani F, Negri F. Microsatellite instability in colorectal cancer. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:97-101. [PMID: 30561401 PMCID: PMC6502181 DOI: 10.23750/abm.v89i9-s.7960] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Indexed: 12/12/2022]
Abstract
Microsatellites are short tandem repeat DNA sequences of one to tetra base pairs distributed throughout the human genome, both in coding and non-coding regions. Owing to their repeated structure, microsatellites are particularly prone to replication errors that are normally repaired by the Mismatch Repair (MMR) system. MMR is a very highly conserved cellular process, involving many proteins, resulting in the identification, and subsequent repair of mismatched bases, likely to have arisen during DNA replication, genetic recombination or chemical or physical damage. Proteins within the MMR system include MLH1, PMS2, MSH2, MSH6, MLH3, MSH3, PMS1, and Exo1. Deficient MMR (dMMR) results in a strong mutator phenotype known as microsatellite instability (MSI), characterized by widespread length polymorphisms of microsatellite sequences due to DNA polymerase slippage. MSI is recognized as one of the major carcinogenetic pathways of colorectal cancer (CRC): it represents a molecular hallmark of hereditary nonpolyposis colorectal cancer (HNPCC), also known as Lynch syndrome (LS); moreover it is detected in 15% of sporadic colorectal cancers, more often due to an epigenetic inactivation of MLH1. Identification of MSI CRC is important, as MSI may serve as a screening tool for detecting LS, a prognostic marker for patient outcome, and a predictive marker for response to chemotherapy and to immunotherapy. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Gian Luigi De' Angelis
- Gastroenterology and Endoscopy Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Mushtaq MU, Papadas A, Pagenkopf A, Flietner E, Morrow Z, Chaudhary SG, Asimakopoulos F. Tumor matrix remodeling and novel immunotherapies: the promise of matrix-derived immune biomarkers. J Immunother Cancer 2018; 6:65. [PMID: 29970158 PMCID: PMC6029413 DOI: 10.1186/s40425-018-0376-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/15/2018] [Indexed: 02/08/2023] Open
Abstract
Recent advances in our understanding of the dynamics of cellular cross-talk have highlighted the significance of host-versus-tumor effect that can be harnessed with immune therapies. Tumors exploit immune checkpoints to evade adaptive immune responses. Cancer immunotherapy has witnessed a revolution in the past decade with the development of immune checkpoint inhibitors (ICIs), monoclonal antibodies against cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) or their ligands, such as PD1 ligand 1 (PD-L1). ICIs have been reported to have activity against a broad range of tumor types, in both solid organ and hematologic malignancy contexts. However, less than one-third of the patients achieve a durable and meaningful treatment response. Expression of immune checkpoint ligands (e.g., PD-L1), mutational burden and tumor-infiltrating lymphocytes are currently used as biomarkers for predicting response to ICIs. However, they do not reliably predict which patients will benefit from these therapies. There is dire need to discover novel biomarkers to predict treatment efficacy and to identify areas for development of combination strategies to improve response rates. Emerging evidence suggests key roles of tumor extracellular matrix (ECM) components and their proteolytic remodeling products in regulating each step of the cancer-immunity cycle. Here we review tumor matrix dynamics and matrix remodeling in context of anti-tumor immune responses and immunotherapy and propose the exploration of matrix-based biomarkers to identify candidates for immune therapy.
Collapse
Affiliation(s)
- Muhammad Umair Mushtaq
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Athanasios Papadas
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Adam Pagenkopf
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Evan Flietner
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Zachary Morrow
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Sibgha Gull Chaudhary
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Fotis Asimakopoulos
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA. .,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA.
| |
Collapse
|
25
|
Alpert L, Pai RK, Srivastava A, McKinnon W, Wilcox R, Yantiss RK, Arcega R, Wang HL, Robert ME, Liu X, Pai RK, Zhao L, Westerhoff M, Hampel H, Kupfer S, Setia N, Xiao SY, Hart J, Frankel WL. Colorectal Carcinomas With Isolated Loss of PMS2 Staining by Immunohistochemistry. Arch Pathol Lab Med 2018; 142:523-528. [DOI: 10.5858/arpa.2017-0156-oa] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Isolated loss of PMS2 staining is an uncommon immunophenotype in colorectal carcinomas, accounting for approximately 4% of tumors with microsatellite instability. Limited information regarding these tumors is available in the literature.
Objective.—
To compare the clinicopathologic features of colorectal carcinomas with isolated PMS2 loss by immunohistochemistry to those with other forms of mismatch repair deficiency.
Design.—
Ninety-three colorectal carcinomas with isolated PMS2 loss by immunohistochemistry and 193 with other forms of mismatch repair deficiency were identified. Forty (43%) of the isolated PMS2 loss cases and 35 control cases (18%) had a known germline mutation or a clinical diagnosis of Lynch syndrome.
Results.—
Overall, isolated PMS2-loss tumors occurred in significantly younger patients (P < .001) and in fewer female patients (P = .006). These tumors were significantly less likely to be right-sided (P = .001), high-grade (P = .01), or display histologic features of microsatellite instability (P < .001). The isolated PMS2-loss group also exhibited increased odds of disease-specific death (odds ratio [OR], 3.09; 95% CI, 1.41–6.85; P = .007). When the analysis was restricted to germline mutation/Lynch syndrome cases and controls, no significant differences were detected for age, sex, tumor location, tumor grade, histologic features, or distant metastases, although a trend toward increased odds of disease-specific death in the isolated PMS2-loss group was evident (OR, 3.87; 95% CI, 0.89–27.04; P = .10).
Conclusions.—
Unusual clinicopathologic features observed in colorectal carcinomas with isolated PMS2 loss are likely related to the high proportion of cases caused by germline mutations. Isolated PMS2-loss tumors may demonstrate more aggressive behavior than other tumors with microsatellite instability, but larger studies are needed to investigate that possibility further.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Wendy L. Frankel
- From the Departments of Pathology (Drs Alpert, Setia, Xiao, and Hart) and Medicine (Dr Kupfer), University of Chicago, Chicago, Illinois; the Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (Dr Reetesh Pai); the Department of Pathology, Brigham and Womens Hospital, Boston, Massachusetts (Dr Srivastava); the Departments of Medicine (Ms McKinnon) and Patho
| |
Collapse
|
26
|
Binder H, Hopp L, Schweiger MR, Hoffmann S, Jühling F, Kerick M, Timmermann B, Siebert S, Grimm C, Nersisyan L, Arakelyan A, Herberg M, Buske P, Loeffler-Wirth H, Rosolowski M, Engel C, Przybilla J, Peifer M, Friedrichs N, Moeslein G, Odenthal M, Hussong M, Peters S, Holzapfel S, Nattermann J, Hueneburg R, Schmiegel W, Royer-Pokora B, Aretz S, Kloth M, Kloor M, Buettner R, Galle J, Loeffler M. Genomic and transcriptomic heterogeneity of colorectal tumours arising in Lynch syndrome. J Pathol 2017; 243:242-254. [DOI: 10.1002/path.4948] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/01/2017] [Accepted: 07/14/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Hans Binder
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Lydia Hopp
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Michal R Schweiger
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Steve Hoffmann
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Frank Jühling
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques; Strasbourg France
- Université de Strasbourg; Strasbourg France
| | - Martin Kerick
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | | | - Susann Siebert
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Christina Grimm
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Lilit Nersisyan
- Group of Bioinformatics, Institute of Molecular Biology; National Academy of Sciences; Yerevan Armenia
| | - Arsen Arakelyan
- Group of Bioinformatics, Institute of Molecular Biology; National Academy of Sciences; Yerevan Armenia
| | - Maria Herberg
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Peter Buske
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Maciej Rosolowski
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University; Leipzig Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University; Leipzig Germany
| | - Jens Przybilla
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Martin Peifer
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Nicolaus Friedrichs
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Gabriela Moeslein
- Department of Hereditary Tumour Syndromes; Surgical Centre, HELIOS Clinic, University Witten/Herdecke; Wuppertal Germany
| | - Margarete Odenthal
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Michelle Hussong
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Sophia Peters
- Institute of Human Genetics, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Stefanie Holzapfel
- Institute of Human Genetics, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Robert Hueneburg
- Department of Internal Medicine I, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Wolff Schmiegel
- Department of Medicine, Haematology and Oncology; Ruhr-University of Bochum, Knappschaftskrankenhaus; Bochum Germany
| | - Brigitte Royer-Pokora
- Institute of Human Genetics and Anthropology; Heinrich-Heine University; Düsseldorf Germany
| | - Stefan Aretz
- Institute of Human Genetics, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Michael Kloth
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology; University Hospital Heidelberg; Heidelberg Germany
- Clinical Cooperation Unit of Applied Tumour Biology; DKFZ (German Cancer Research Centre) Heidelberg; Germany
- Molecular Medicine Partnership Unit; University Hospital Heidelberg and EMBL Heidelberg; Heidelberg Germany
| | - Reinhard Buettner
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Jörg Galle
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University; Leipzig Germany
| |
Collapse
|
27
|
Inderberg EM, Wälchli S, Myhre MR, Trachsel S, Almåsbak H, Kvalheim G, Gaudernack G. T cell therapy targeting a public neoantigen in microsatellite instable colon cancer reduces in vivo tumor growth. Oncoimmunology 2017; 6:e1302631. [PMID: 28507809 PMCID: PMC5414866 DOI: 10.1080/2162402x.2017.1302631] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 02/08/2023] Open
Abstract
T-cell receptor (TCR) transfer is an attractive strategy to increase the number of cancer-specific T cells in adoptive cell therapy. However, recent clinical and pre-clinical findings indicate that careful consideration of the target antigen is required to limit the risk of off-target toxicity. Directing T cells against mutated proteins such as frequently occurring frameshift mutations may thus be a safer alternative to tumor-associated self-antigens. Furthermore, such frameshift mutations result in novel polypeptides allowing selection of TCRs from the non-tolerant T-cell repertoire circumventing the problem of low affinity TCRs due to central tolerance. The transforming growth factor β Receptor II frameshift mutation (TGFβRIImut) is found in Lynch syndrome cancer patients and in approximately 15% of sporadic colorectal and gastric cancers displaying microsatellite instability (MSI). The -1A mutation within a stretch of 10 adenine bases (nucleotides 709-718) of the TGFβRII gene gives rise to immunogenic peptides previously used for vaccination of MSI+ colorectal cancer patients in a Phase I clinical trial. From a clinically responding patient, we isolated a cytotoxic T lymphocyte (CTL) clone showing a restriction for HLA-A2 in complex with TGFβRIImut peptide. Its TCR was identified and shown to redirect T cells against colon carcinoma cell lines harboring the frameshift mutation. Finally, T cells transduced with the HLA-A2-restricted TGFβRIImut-specific TCR were demonstrated to significantly reduce the growth of colorectal cancer and enhance survival in a NOD/SCID xenograft mouse model.
Collapse
Affiliation(s)
- Else M Inderberg
- Section for Cellular Therapy, Department for Cancer Treatment, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Section for Cellular Therapy, Department for Cancer Treatment, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Section for Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Marit R Myhre
- Section for Cellular Therapy, Department for Cancer Treatment, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Sissel Trachsel
- Section for Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Hilde Almåsbak
- Section for Cellular Therapy, Department for Cancer Treatment, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Gunnar Kvalheim
- Section for Cellular Therapy, Department for Cancer Treatment, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Gustav Gaudernack
- Section for Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
28
|
Duruisseaux M, Lize-Dufranc C, Badoual C, Bibeau F. Biomarqueurs prédictifs de l’efficacité des inhibiteurs de checkpoint immunitaire dans le traitement des cancers. Ann Pathol 2017; 37:46-54. [DOI: 10.1016/j.annpat.2016.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/14/2016] [Indexed: 12/25/2022]
|
29
|
A Nomogram to Predict Adequate Lymph Node Recovery before Resection of Colorectal Cancer. PLoS One 2016; 11:e0168156. [PMID: 27992611 PMCID: PMC5161509 DOI: 10.1371/journal.pone.0168156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/25/2016] [Indexed: 12/15/2022] Open
Abstract
Increased lymph node count (LNC) has been associated with prolonged survival in colorectal cancer (CRC), but the underlying mechanisms are still poorly understood. The study aims to identify new predictors and develop a preoperative nomogram for predicting the probability of adequate LNC (≥ 12). 501 eligible patients were retrospectively selected to identify clinical-pathological factors associated with LNC ≥ 12 through univariate and multivariate logistic regression analyses. The nomogram was built according to multivariate analyses of preoperative factors. Model performance was assessed with concordance index (c-index) and area under the receiver operating characteristic curve (AUC), followed by internal validation and calibration using 1000-resample bootstrapping. Clinical validity of the nomogram and LNC impact on stage migration were also evaluated. Multivariate analyses showed patient age, CA19-9, circulating lymphocytes, neutrophils, platelets, tumor diameter, histology and deposit significantly correlated with LNC (P < 0.05). The effects were marginal for CEA, anemia and CRC location (0.05 < P < 0.1). The multivariate analyses of preoperative factors suggested decreased age, CEA, CA19-9, neutrophils, proximal location, and increased platelets and diameter were significantly associated with increased probability of LNC ≥ 12 (P < 0.05). The nomogram achieved c-indexes of 0.75 and 0.73 before and after correction for overfitting. The AUC was 0.75 (95% CI, 0.70–0.79) and the clinically valid threshold probabilities were between 10% and 60% for the nomogram to predict LNC < 12. Additionally, increased probability of adequate LNC before surgery was associated with increased LNC and negative lymph nodes rather than increased positive lymph nodes, lymph node ratio, pN stages or AJCC stages. Collectively, the results indicate the LNC is multifactorial and irrelevant to stage migration. The significant correlations with preoperative circulating markers may provide new explanations for LNC-related survival advantage which is reflected by the implication of regional and systemic antitumor immune responses.
Collapse
|
30
|
Patients with genetically heterogeneous synchronous colorectal cancer carry rare damaging germline mutations in immune-related genes. Nat Commun 2016; 7:12072. [PMID: 27377421 PMCID: PMC4935966 DOI: 10.1038/ncomms12072] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/26/2016] [Indexed: 12/16/2022] Open
Abstract
Synchronous colorectal cancers (syCRCs) are physically separated tumours that develop simultaneously. To understand how the genetic and environmental background influences the development of multiple tumours, here we conduct a comparative analysis of 20 syCRCs from 10 patients. We show that syCRCs have independent genetic origins, acquire dissimilar somatic alterations, and have different clone composition. This inter- and intratumour heterogeneity must be considered in the selection of therapy and in the monitoring of resistance. SyCRC patients show a higher occurrence of inherited damaging mutations in immune-related genes compared to patients with solitary colorectal cancer and to healthy individuals from the 1,000 Genomes Project. Moreover, they have a different composition of immune cell populations in tumour and normal mucosa, and transcriptional differences in immune-related biological processes. This suggests an environmental field effect that promotes multiple tumours likely in the background of inflammation. Some individuals present with multiple synchronous colorectal tumours, but the genetic understanding of this is unclear. Here, the authors use a sequencing strategy to show that the synchronous tumours are genetically independent and the patients harbour rare germline damaging mutations in genes associated with the immune system.
Collapse
|
31
|
McMeekin DS, Tritchler DL, Cohn DE, Mutch DG, Lankes HA, Geller MA, Powell MA, Backes FJ, Landrum LM, Zaino R, Broaddus RD, Ramirez N, Gao F, Ali S, Darcy KM, Pearl ML, DiSilvestro PA, Lele SB, Goodfellow PJ. Clinicopathologic Significance of Mismatch Repair Defects in Endometrial Cancer: An NRG Oncology/Gynecologic Oncology Group Study. J Clin Oncol 2016; 34:3062-8. [PMID: 27325856 DOI: 10.1200/jco.2016.67.8722] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE The clinicopathologic significance of mismatch repair (MMR) defects in endometrioid endometrial cancer (EEC) has not been definitively established. We undertook tumor typing to classify MMR defects to determine if MMR status is prognostic or predictive. METHODS Primary EECs from NRG/GOG0210 patients were assessed for microsatellite instability (MSI), MLH1 methylation, and MMR protein expression. Each tumor was assigned to one of four MMR classes: normal, epigenetic defect, probable mutation (MMR defect not attributable to MLH1 methylation), or MSI-low. The relationships between MMR classes and clinicopathologic variables were assessed using contingency table tests and Cox proportional hazard models. RESULTS A total of 1,024 tumors were assigned to MMR classes. Epigenetic and probable mutations in MMR were significantly associated with higher grade and more frequent lymphovascular space invasion. Epigenetic defects were more common in patients with higher International Federation of Gynecology and Obstetrics stage. Overall, there were no differences in outcomes. Progression-free survival was, however, worse for women whose tumors had epigenetic MMR defects compared with the MMR normal group (hazard ratio, 1.37; P < .05; 95% CI, 1.00 to 1.86). An exploratory analysis of interaction between MMR status and adjuvant therapy showed a trend toward improved progression-free survival for probable MMR mutation cases. CONCLUSION MMR defects in EECs are associated with a number of well-established poor prognostic indicators. Women with tumors that had MMR defects were likely to have higher-grade cancers and more frequent lymphovascular space invasion. Surprisingly, outcomes in these patients were similar to patients with MMR normal tumors, suggesting that MMR defects may counteract the effects of negative prognostic factors. Altered immune surveillance of MMR-deficient tumors, and other host/tumor interactions, is likely to determine outcomes for patients with MMR-deficient tumors.
Collapse
Affiliation(s)
- D Scott McMeekin
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - David L Tritchler
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - David E Cohn
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - David G Mutch
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Heather A Lankes
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Melissa A Geller
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Matthew A Powell
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Floor J Backes
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Lisa M Landrum
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Richard Zaino
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Russell D Broaddus
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Nilsa Ramirez
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Feng Gao
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Shamshad Ali
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Kathleen M Darcy
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Michael L Pearl
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Paul A DiSilvestro
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Shashikant B Lele
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Paul J Goodfellow
- D. Scott McMeekin and Lisa M. Landrum, University of Oklahoma Health Sciences Center, Oklahoma City, OK; David L. Tritchler, Heather A. Lankes, and Shamshad Ali, NRG Oncology Statistics and Data Management Center; Shashikant B. Lele, Roswell Park Cancer Institute, Buffalo; Michael L. Pearl, Stony Brook University Hospital, Stony Brook, NY; David E. Cohn, Floor J. Backes, and Paul J. Goodfellow, The Ohio State University; Nilsa Ramirez, Research Institute at Nationwide Children's Hospital, Columbus, OH; David G. Mutch, Matthew A. Powell, and Feng Gao, Washington University School of Medicine, St. Louis, MO; Melissa A. Geller, University of Minnesota, Minneapolis, MN; Richard Zaino, Penn State Milton S. Hersey Medical Center, Hershey, PA; Russell D. Broaddus, The University of Texas MD Anderson Cancer Center, Houston, TX; Kathleen M. Darcy, Women's Health Integrated Research Collective, Annandale, VA; and Paul A. DiSilvestro, Women and Infants Hospital of Rhode Island, Providence, RI.
| |
Collapse
|
32
|
Rozek LS, Schmit SL, Greenson JK, Tomsho LP, Rennert HS, Rennert G, Gruber SB. Tumor-Infiltrating Lymphocytes, Crohn's-Like Lymphoid Reaction, and Survival From Colorectal Cancer. J Natl Cancer Inst 2016; 108:djw027. [PMID: 27172903 DOI: 10.1093/jnci/djw027] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 02/05/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND While clinical outcomes from colorectal cancer (CRC) are influenced by stage at diagnosis and treatment, mounting evidence suggests that an enhanced lymphocytic reaction to a tumor may also be an informative prognostic indicator. METHODS The roles of intratumoral T lymphocyte infiltration (TIL), peritumoral Crohn's-like lymphoid reaction (CLR), microsatellite instability (MSI), and clinicopathological characteristics in survival from CRC were examined using 2369 incident CRCs from a population-based case-control study in northern Israel. Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for CRC-specific and all-cause mortality in multivariable models adjusted for age, sex, ethnicity, grade, stage, and MSI. All statistical tests were two-sided. RESULTS Tumors with TIL/high-powered field (HPF) of 2 or greater were associated with a statistically significant increase in CRC-specific (P < .001) and overall survival (P < .001) compared with tumors with TIL/HPF of less than 2. Similarly, tumors with a prominent CLR experienced better CRC-specific (P < .001) and overall survival (P < .001) as compared with those with no response. High TILs (HR = 0.76, 95% CI = 0.64 to 0.89, P < .001) and a prominent CLR (HR = 0.71, 95% CI = 0.62 to 0.80, P < .001), but not MSI, were associated with a statistically significant reduction in all-cause mortality after adjustment for established prognostic factors. CONCLUSIONS TILs and CLR are both prognostic indicators for CRC after adjusting for traditional prognostic indicators.
Collapse
Affiliation(s)
- Laura S Rozek
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| | - Stephanie L Schmit
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| | - Joel K Greenson
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| | - Lynn P Tomsho
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| | - Hedy S Rennert
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| | - Gad Rennert
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| | - Stephen B Gruber
- Affiliations of authors: Department of Environmental Health Sciences, University of Michigan School of Public Health (LSR), and Department of Pathology (JKG) and Department of Internal Medicine (LPT), University of Michigan Medical School, Ann Arbor, MI; USC Norris Comprehensive Cancer Center and Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA (SLS, SBG); Clalit National Israeli Cancer Control Center, Haifa, Israel (HSR, GR)
| |
Collapse
|
33
|
Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016; 16:275-87. [PMID: 27079802 PMCID: PMC5381938 DOI: 10.1038/nrc.2016.36] [Citation(s) in RCA: 1985] [Impact Index Per Article: 220.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
With recent approvals for multiple therapeutic antibodies that block cytotoxic T lymphocyte associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) in melanoma, non-small-cell lung cancer and kidney cancer, and additional immune checkpoints being targeted clinically, many questions still remain regarding the optimal use of drugs that block these checkpoint pathways. Defining biomarkers that predict therapeutic effects and adverse events is a crucial mandate, highlighted by recent approvals for two PDL1 diagnostic tests. Here, we discuss biomarkers for anti-PD1 therapy based on immunological, genetic and virological criteria. The unique biology of the CTLA4 immune checkpoint, compared with PD1, requires a different approach to biomarker development. Mechanism-based insights from such studies may guide the design of synergistic treatment combinations based on immune checkpoint blockade.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| | - Robert A Anders
- Department of Pathology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| |
Collapse
|
34
|
Pine JK, Morris E, Hutchins GG, West NP, Jayne DG, Quirke P, Prasad KR. Systemic neutrophil-to-lymphocyte ratio in colorectal cancer: the relationship to patient survival, tumour biology and local lymphocytic response to tumour. Br J Cancer 2015; 113:204-11. [PMID: 26125452 PMCID: PMC4506398 DOI: 10.1038/bjc.2015.87] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/08/2014] [Accepted: 01/15/2015] [Indexed: 12/19/2022] Open
Abstract
Background: Colorectal cancer (CRC) is a major cause of mortality and morbidity. The impact of inflammatory biomarkers (C-reactive protein etc.) on CRC is increasingly studied including systemic neutrophil-to-lymphocyte ratio (NLR) as they seem to predict outcome. Methods: All patients who underwent curative resection for CRC from 2000 to 2004 at Leeds Teaching Hospitals NHS Trust had pre-operative NLR calculated. Demographic, histopathological and survival data were collected. Tissue microarrays were created and stained to determine the mismatch repair (MMR) protein status of each tumour. Local lymphocytic response to the tumour was assessed and graded. Results: About 358 patients were eligible. Of these 88 had an NLR ⩾5, which predicted lower overall survival and greater disease recurrence. A high NLR is associated with higher pT- and pN-stage and a greater incidence of extramural venous invasion. MMR protein status was not associated with NLR. A pronounced lymphocytic reaction at the invasive margin (IM) indicated a better prognosis and was associated with a lower NLR. Conclusion: Neutrophil-to-lymphocyte ratio predicts disease-free and overall survival and is associated with a more aggressive tumour phenotype. The lymphocytic response to tumour at the IM is associated with NLR however dMMR is not. Neutrophil-to-lymphocyte ratio is a cheap, easy-to-access test that predicts outcome in CRC.
Collapse
Affiliation(s)
- J K Pine
- 1] Department of HPB Surgery, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK [2] Department of Pathology and Tumour Biology, Leeds Institute of Cancer and Pathology, University of Leeds LS9 7TF, Leeds, UK
| | - E Morris
- Cancer Epidemiology Group, Leeds Institute of Cancer and Pathology, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - G G Hutchins
- Department of Pathology and Tumour Biology, Leeds Institute of Cancer and Pathology, University of Leeds LS9 7TF, Leeds, UK
| | - N P West
- Department of Pathology and Tumour Biology, Leeds Institute of Cancer and Pathology, University of Leeds LS9 7TF, Leeds, UK
| | - D G Jayne
- Department of Colorectal Surgery, Leeds Institute of Biomedical & Clinical Sciences, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - P Quirke
- Department of Pathology and Tumour Biology, Leeds Institute of Cancer and Pathology, University of Leeds LS9 7TF, Leeds, UK
| | - K R Prasad
- Department of HPB Surgery, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| |
Collapse
|
35
|
Microsatellite instability, disease-free survival and role of tumour infiltrating lymphocytes (Invited editorial on ‘Predictors of disease-free survival in colorectal cancer with microsatellite instability: An AGEO multicenter study’). Eur J Cancer 2015; 51:922-4. [DOI: 10.1016/j.ejca.2015.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/16/2015] [Indexed: 01/30/2023]
|
36
|
Nikulina VV, Garmanchuk LV, Senchylo NV, Nikolaenko TV, Dzhus OI, Ostapchenko LI, Khranovska NM. Combined influence of teichoic acids from Staphylococcus aureus and heterometallik Cu/Cd ethylenediamine complex on peritoneal macrophages and tumor cells. CYTOL GENET+ 2014. [DOI: 10.3103/s0095452714060085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
37
|
Mismatch repair protein deficiency is common in sebaceous neoplasms and suggests the importance of screening for Lynch syndrome. Am J Dermatopathol 2013; 35:191-5. [PMID: 22722469 DOI: 10.1097/dad.0b013e31825f7efe] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The association between Lynch syndrome and sebaceous neoplasms is well characterized. The absence of expression of mismatch repair proteins (MMRPs) by immunohistochemistry (IHC) is often used in other Lynch-associated tumors to guide testing. IHC for MLH1, PMS2, MSH2, and MSH6 was performed on 36 benign and malignant sebaceous neoplasms with the absence of one or more MMRP in 38.9% of cases. Among lesions with abnormal IHC, 71.4% were missing both MSH2 and MSH6, 21.4% lacked MLH1 and PMS2, and 7.1% lacked only MSH6. Of the 10 patients with absent MMRP, 5 had gene-test confirmed Lynch syndrome, 3 had no suggestive personal or family medical history and 2 had no recorded data. Tumor-infiltrating lymphocytes in neoplasms with absent MMRP were statistically significantly greater than in those with intact MMRP (16.5 vs. 9.7, P = 0.027). MMRP deficiency is common in sebaceous neoplasms, suggesting the importance of screening for Lynch syndrome in these patients.
Collapse
|
38
|
Tougeron D, Maby P, Elie N, Fauquembergue E, Le Pessot F, Cornic M, Sabourin JC, Michel P, Frébourg T, Latouche JB. Regulatory T lymphocytes are associated with less aggressive histologic features in microsatellite-unstable colorectal cancers. PLoS One 2013; 8:e61001. [PMID: 23613769 PMCID: PMC3626697 DOI: 10.1371/journal.pone.0061001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/05/2013] [Indexed: 12/21/2022] Open
Abstract
Background Colorectal cancers (CRCs) with microsatellite instability (MSI) are associated with a good prognosis and a high density of tumor-infiltrating lymphocytes (TILs). We have undertaken to determine the link between TIL densities and MSI CRC histologic features. Patients and Methods Using tissue microarrays, T-cell sub-population infiltration, including T cells (CD3), cytotoxic T cells (CD8) and regulatory T cells (FoxP3) were studied in 86 MSI CRCs. We separately analyzed TILs of the stromal and epithelial compartments in the tumor center, the tumoral invasion margin and associated normal tissue. Results For FoxP3+ TIL density in the tumor center stromal compartment, we found a strong negative correlation with T4 stage (p = 0.01), node invasion (p<0.001) and VELIPI (vascular emboli, lymphatic invasion and perinervous invasion) criteria (p = 0.002). Conclusion The strong correlation between regulatory T cell density and the absence of VELIPI criteria suggests that this sub-group of T cells is preferentially associated with less invasive tumors.
Collapse
Affiliation(s)
- David Tougeron
- Inserm, U1079, Faculty of Medicine, Institute for Medical Research, Rouen University Hospital, Rouen, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gavin PG, Colangelo LH, Fumagalli D, Tanaka N, Remillard MY, Yothers G, Kim C, Taniyama Y, Kim SI, Choi HJ, Blackmon NL, Lipchik C, Petrelli NJ, O'Connell MJ, Wolmark N, Paik S, Pogue-Geile KL. Mutation profiling and microsatellite instability in stage II and III colon cancer: an assessment of their prognostic and oxaliplatin predictive value. Clin Cancer Res 2012; 18:6531-41. [PMID: 23045248 DOI: 10.1158/1078-0432.ccr-12-0605] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE The purpose of this study was to examine the prognostic and oxaliplatin predictive value of mismatch repair (MMR) status and common hot spot mutations, which we previously identified in stage II and III colon cancer. EXPERIMENTAL DESIGN Mutations in BRAF, KRAS, NRAS, MET, and PIK3CA were profiled in 2,299 stage II and III colon tumors from National Surgical Adjuvant Breast and Bowel Project (NSABP) clinical trials C-07 (n = 1,836) and C-08 (n = 463) with Type Plex chemistry and mass spectrometry. C-07 tested the worth of adding oxaliplatin to 5-fluorouracil plus leucovorin, and C-08 tested the worth of adding bevacizumab to FOLFOX. Cox proportional hazard models were used to assess prognostic or oxaliplatin predictive value of mutations for tumor recurrence, overall survival (OS), and survival after recurrence (SAR). RESULTS BRAF mutations were associated with MMR-deficient tumors (P < 0.0001), poor OS [HR, 1.46; 95% confidence interval (CI), 1.20-1.79; P ≤ 0.0002], and poor SAR (HR, 2.31; 95% CI, 1.83-2.95; P < 0.0001). Mutations in KRAS, NRAS, MET, and PIK3CA were not associated with recurrence, OS, or SAR. MMR-deficient tumors were associated with an improved prognosis based on recurrence (HR, 0.48; 95% CI, 0.33-0.70; P < 0.0001). Mutations and MMR status were not predictive for oxaliplatin benefit. CONCLUSIONS This study shows that BRAF mutations profiled from stage II and III colon cancer tumors were associated with poor SAR and validates and explains, at least in part, previous observations associating it with poor OS. Profiling of all of these mutations is warranted for future clinical trials testing new targeted therapies that block relevant signaling pathways. Such clinical trials are under development at NSABP.
Collapse
Affiliation(s)
- Patrick G Gavin
- National Surgical Adjuvant Breast and Bowel Project, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15212, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bozzao C, Lastella P, Stella A. Anticipation in lynch syndrome: where we are where we go. Curr Genomics 2012; 12:451-65. [PMID: 22547953 PMCID: PMC3219841 DOI: 10.2174/138920211797904070] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/08/2011] [Accepted: 07/20/2011] [Indexed: 02/06/2023] Open
Abstract
Lynch syndrome (LS) is the most common form of inherited predisposition to develop cancer mainly in the colon and endometrium but also in other organ sites. Germline mutations in DNA mismatch repair (MMR) gene cause the transmission of the syndrome in an autosomal dominant manner. The management of LS patients is complicated by the large variation in age at cancer diagnosis which requires these patients to be enrolled in surveillance protocol starting as early as in their second decade of life. Several environmental and genetic factors have been proposed to explain this phenotypic heterogeneity, but the molecular mechanisms remain unknown. Although the presence of genetic anticipation in Lynch syndrome has been suspected since 15 years, only recently the phenomenon has been increasingly reported to be present in different cancer genetic syndromes including LS. While the biological basis of earlier cancer onset in successive generations remains poorly known, recent findings point to telomere dynamics as a mechanism significantly contributing to genetic anticipation in Lynch syndrome and in other familial cancers. In this review, we summarize the clinical and molecular features of Lynch syndrome, with a particular focus on the latest studies that have investigated the molecular mechanisms of genetic anticipation.
Collapse
Affiliation(s)
- Cristina Bozzao
- Medical Genetics Unit, Department of Biomedicine in Childhood, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | | | | |
Collapse
|
41
|
Deschoolmeester V, Baay M, Lardon F, Pauwels P, Peeters M. Immune Cells in Colorectal Cancer: Prognostic Relevance and Role of MSI. CANCER MICROENVIRONMENT 2011; 4:377-92. [PMID: 21618031 DOI: 10.1007/s12307-011-0068-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 05/19/2011] [Indexed: 12/14/2022]
Abstract
There is growing evidence that both local and systemic inflammatory responses play an important role in the progression of a variety of solid tumors. Colorectal cancer (CRC) results from the cumulative effect of sequential genetic alterations, leading to the expression of tumor-associated antigens possibly inducing a cellular anti-tumor immune response. It is well recognized that cytotoxic lymphocytes (CTLs) constitute one of the most important effector mechanisms of anti-tumor-immunity. However, their potential prognostic influence in CRC remains controversial. In addition, other key players like natural killer cells, tumor associated macrophages and regulatory T cells play an important role in the immune attack against CRC and need further investigation. This review will mainly focus on the role of the adaptive immune system in CRC and particularly in regard to microsatellite instability.
Collapse
Affiliation(s)
- Vanessa Deschoolmeester
- Laboratory of Cancer Research and Clinical Oncology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium,
| | | | | | | | | |
Collapse
|
42
|
Abstract
Microsatellite instability (MSI) occurs in about 15% of gastrointestinal cancers and it is associated with specific clinic, pathologic, and molecular features of the tumors. MSI-high (MSI-H) carcinomas also follow specific tumor development pathways. This review is focused on the molecular profile of alterations in members of the KRAS signaling pathway (EGFR, KRAS, BRAF, PIK3CA, RASSF1A, and MLK3 genes) in MSI gastrointestinal carcinomas. Alterations in these genes characterize more than half of gastrointestinal cancers and frequently occur simultaneously in the same tumor, pinpointing the KRAS signaling pathway as one of the most frequently altered pathways in this subset of cancers. Nowadays, many and novel inhibitors targeting molecules of this signaling pathway are being described; therefore, it is worthwhile to test their efficacy in MSI gastrointestinal cancers in order to develop new and more directed targeted therapies for patients affected by this disease.
Collapse
|
43
|
Duggan S, Prichard D, Kirca M, Kelleher D. Inherited Syndromes Predisposing to Inflammation and GI Cancer. Recent Results Cancer Res 2011; 185:35-50. [PMID: 21822818 DOI: 10.1007/978-3-642-03503-6_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancers arising within the gastrointestinal (GI) tract are commonly associated with an immune component at their inception and later in their maintenance. While many of the immune factors and immune cell types surrounding these lesions have been highlighted, the underlying pre-dispositions in immunesupported carcinogenesis are not well characterised. Inherited Mendelian GI disorders such as polyposis syndromes, while classically due to germline mutations in non-immune genes, commonly demonstrate alterations in key immune and inflammatory genes. In some cases immune based therapies have been shown to provide at least some benefit in animal models of these syndromes. The advent of genome wide association studies has begun to powerfully examine the genetic nature of complex non-Mendelian GI diseases highlighting polymorphisms within immune related genes and their potential to provide the niche in which GI cancers may originate. Here in the role in which Mendelian and non-Mendelian genetics of immune related factors supporting GI malignancy will be presented and discussed.
Collapse
Affiliation(s)
- Shane Duggan
- Department of Clinical Medicine and Institute of Molecular Medicine, Trinity College Dublin, Ireland
| | | | | | | |
Collapse
|
44
|
Deschoolmeester V, Baay M, Specenier P, Lardon F, Vermorken JB. A review of the most promising biomarkers in colorectal cancer: one step closer to targeted therapy. Oncologist 2010; 15:699-731. [PMID: 20584808 PMCID: PMC3228001 DOI: 10.1634/theoncologist.2010-0025] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 05/01/2010] [Indexed: 02/06/2023] Open
Abstract
Rapidly growing insights into the molecular biology of colorectal cancer (CRC) and recent developments in gene sequencing and molecular diagnostics have led to high expectations for the identification of molecular markers to be used in optimized and tailored treatment regimens. However, many of the published data on molecular biomarkers are contradictory in their findings and the current reality is that no molecular marker, other than the KRAS gene in the case of epidermal growth factor receptor (EGFR)- targeted therapy for metastatic disease, has made it into clinical practice. Many markers investigated suffer from technical shortcomings, resulting from lack of quantitative techniques to capture the impact of the molecular alteration. This understanding has recently led to the more comprehensive approaches of global gene expression profiling or genome-wide analysis to determine prognostic and predictive signatures in tumors. In this review, an update of the most recent data on promising biological prognostic and/or predictive markers, including microsatellite instability, epidermal growth factor receptor, KRAS, BRAF, CpG island methylator phenotype, cytotoxic T lymphocytes, forkhead box P3-positive T cells, receptor for hyaluronic acid-mediated motility, phosphatase and tensin homolog, and T-cell originated protein kinase, in patients with CRC is provided.
Collapse
Affiliation(s)
- Vanessa Deschoolmeester
- Laboratory of Cancer Research and Clinical Oncology, Department of Medical Oncology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | | | | | | | | |
Collapse
|
45
|
Drescher KM, Sharma P, Lynch HT. Current hypotheses on how microsatellite instability leads to enhanced survival of Lynch Syndrome patients. Clin Dev Immunol 2010; 2010:170432. [PMID: 20631828 PMCID: PMC2901607 DOI: 10.1155/2010/170432] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 04/13/2010] [Indexed: 01/20/2023]
Abstract
High levels of microsatellite instability (MSI-high) are a cardinal feature of colorectal tumors from patients with Lynch Syndrome. Other key characteristics of Lynch Syndrome are that these patients experience fewer metastases and have enhanced survival when compared to patients diagnosed with microsatellite stable (MSS) colorectal cancer. Many of the characteristics associated with Lynch Syndrome including enhanced survival are also observed in patients with sporadic MSI-high colorectal cancer. In this review we will present the current state of knowledge regarding the mechanisms that are utilized by the host to control colorectal cancer in Lynch Syndrome and why these same mechanisms fail in MSS colorectal cancers.
Collapse
Affiliation(s)
- Kristen M Drescher
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA.
| | | | | |
Collapse
|
46
|
Deschoolmeester V, Baay M, Van Marck E, Weyler J, Vermeulen P, Lardon F, Vermorken JB. Tumor infiltrating lymphocytes: an intriguing player in the survival of colorectal cancer patients. BMC Immunol 2010; 11:19. [PMID: 20385003 PMCID: PMC2864219 DOI: 10.1186/1471-2172-11-19] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 04/12/2010] [Indexed: 02/08/2023] Open
Abstract
Background There is growing evidence that both local and systemic inflammatory responses play an important role in the progression of a variety of solid tumors. Colorectal cancer results from the cumulative effect of sequential genetic alterations, leading to the expression of tumor associated antigens possibly inducing a cellular anti-tumor immune response. It is well recognized that cytotoxic lymphocytes constitute one of the most important effector mechanisms of anti-tumor-immunity. However, their potential prognostic influence in colorectal cancer remains controversial. Aim of the study was to examine infiltration of CD3+ and CD8+ lymphocytes in colorectal cancer and their prognostic potential. Two-hundred-fifteen colorectal cancer cases, previously analyzed for microsatellite instability (MSI), were selected for immunohistochemical detection of CD3+, CD8+ infiltration and the expression of granzyme B. Prognostic relevance was assessed by survival analysis. Results Strong correlations were found between the infiltration of lymphocytes and several clinicopathological variables. Survival analysis revealed that intra-epithelial infiltration of CD3+ and CD8+ T lymphocytes and stromal infiltration of CD3+ lymphocytes had a major impact on the patients' overall survival in the univariate analysis, however independent of their association with MSI-status. In addition, it was also demonstrated that there was an important disease specific survival advantage for patients with microsatellite stable (MSS) tumors containing intraepithelial CD8+ tumor infiltrating lymphocytes. When samples were analyzed for colon cancer and rectal cancer separately, the results of the overall population were confirmed in colon cancer only. When entered into a multiple Cox regression analysis adjusting for other possible important confounding factors, the strong impact of lymphocyte infiltration on overall survival was not maintained. Only early stage and young age (borderline significant for overall population only) were associated with a better overall survival (early disease with disease-free survival also). Conclusions In conclusion our results suggest a role for infiltrating CD3+ and CD8+ T lymphocytes in colorectal cancer whereby tumor infiltration could reflect a general principle of antitumor immunity, irrespective of the MSI-status.
Collapse
Affiliation(s)
- Vanessa Deschoolmeester
- Laboratory of Cancer Research and Clinical Oncology, Department of Medical Oncology, University of Antwerp (UA/UZA), Wilrijk, Belgium.
| | | | | | | | | | | | | |
Collapse
|
47
|
Zlobec I, Lugli A. Invasive front of colorectal cancer: Dynamic interface of pro-/anti-tumor factors. World J Gastroenterol 2009; 15:5898-906. [PMID: 20014453 PMCID: PMC2795176 DOI: 10.3748/wjg.15.5898] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tumor-host interaction at the invasive front of colorectal cancer represents a critical interface encompassing a dynamic process of de-differentiation of colorectal carcinoma cells known as epithelial mesenchymal transition (EMT). EMT can be identified histologically by the presence of “tumor budding”, a feature which can be highly specific for tumors showing an infiltrating tumor growth pattern. Importantly, tumor budding and tumor border configuration have generated considerable interest as additional prognostic factors and are also recognized as such by the International Union Against Cancer. Evidence seems to suggest that the presence of tumor budding or an infiltrating growth pattern is inversely correlated with the presence of immune and inflammatory responses at the invasive tumor front. In fact, several tumor-associated antigens such as CD3, CD4, CD8, CD20, Granzyme B, FOXP3 and other immunological or inflammatory cell types have been identified as potentially prognostic in patients with this disease. Evidence seems to suggest that the balance between pro-tumor (including budding and infiltrating growth pattern) and anti-tumor (immune response or certain inflammatory cell types) factors at the invasive front of colorectal cancer may be decisive in determining tumor progression and the clinical outcome of patients with colorectal cancer. On one hand, the infiltrating tumor border configuration and tumor budding promote progression and dissemination of tumor cells by penetrating the vascular and lymphatic vessels. On the other, the host attempts to fend off this attack by mounting an immune response to protect vascular and lymphatic channels from invasion by tumor buds. Whereas standard pathology reporting of breast and prostate cancer involves additional prognostic features, such as the BRE and Gleason scores, the ratio of pro- and anti-tumor factors could be a promising approach for the future development of a prognostic score for patients with colorectal cancer which could complement tumor node metastasis staging to improve the clinical management of patients with this disease.
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW To review data demonstrating the prognostic and predictive impact of microsatellite instability (MSI) in human colon carcinomas. RECENT FINDINGS MSI is a molecular marker of defective DNA mismatch repair that is detected in approximately 15% of sporadic colon cancers. Most, but not all retrospective studies, have shown that colon cancers with MSI have better stage-adjusted survival rates compared with non-MSI tumors. Furthermore, analyses of colon cancers from participants in randomized adjuvant therapy trials have suggested that MSI tumors do not benefit from treatment with 5-fluorouracil. Recent studies, including a pooled analysis, validate prior data demonstrating the prognostic and predictive impact of MSI status in colon cancer. SUMMARY MSI is a molecular marker that can provide valuable prognostic and predictive information in colon cancer patients. In the appropriate clinical setting, MSI data can be used in clinical decision-making. Specifically, the favorable outcome of stage II colon cancers with MSI indicates that such patients should not receive adjuvant chemotherapy. Although data for stage III colon cancers with MSI suggest a lack of benefit from 5-fluorouracil alone, the benefit of the current standard treatment, 5-fluorouracil, leucovorin, and oxaliplatin, in this subgroup remains unknown and awaits further study.
Collapse
|