51
|
Kelly RJ, Bever K, Chao J, Ciombor KK, Eng C, Fakih M, Goyal L, Hubbard J, Iyer R, Kemberling HT, Krishnamurthi S, Ku G, Mordecai MM, Morris VK, Paulson AS, Peterson V, Shah MA, Le DT. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of gastrointestinal cancer. J Immunother Cancer 2023; 11:e006658. [PMID: 37286304 PMCID: PMC10254964 DOI: 10.1136/jitc-2022-006658] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2023] [Indexed: 06/09/2023] Open
Abstract
Gastrointestinal (GI) cancers, including esophageal, gastroesophageal junction, gastric, duodenal and distal small bowel, biliary tract, pancreatic, colon, rectal, and anal cancer, comprise a heterogeneous group of malignancies that impose a significant global burden. Immunotherapy has transformed the treatment landscape for several GI cancers, offering some patients durable responses and prolonged survival. Specifically, immune checkpoint inhibitors (ICIs) directed against programmed cell death protein 1 (PD-1), either as monotherapies or in combination regimens, have gained tissue site-specific regulatory approvals for the treatment of metastatic disease and in the resectable setting. Indications for ICIs in GI cancer, however, have differing biomarker and histology requirements depending on the anatomic site of origin. Furthermore, ICIs are associated with unique toxicity profiles compared with other systemic treatments that have long been the mainstay for GI cancer, such as chemotherapy. With the goal of improving patient care by providing guidance to the oncology community, the Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop this clinical practice guideline on immunotherapy for the treatment of GI cancer. Drawing from published data and clinical experience, the expert panel developed evidence- and consensus-based recommendations for healthcare professionals using ICIs to treat GI cancers, with topics including biomarker testing, therapy selection, and patient education and quality of life considerations, among others.
Collapse
Affiliation(s)
- Ronan J Kelly
- Charles A. Sammons Cancer Center, Baylor University Medical Center at Dallas, Dallas, Texas, USA
| | - Katherine Bever
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph Chao
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Kristen K Ciombor
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Cathy Eng
- Department of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Marwan Fakih
- Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center Duarte, Duarte, California, USA
| | - Lipika Goyal
- Department of Medicine, Stanford University, Palo Alto, California, USA
| | - Joleen Hubbard
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Renuka Iyer
- Department of GI Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Holly T Kemberling
- Department of GI Immunology Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | | | - Geoffrey Ku
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Andrew Scott Paulson
- Department of Medical Oncology, Texas Oncology-Baylor Charles A Sammons Cancer Center, Dallas, Texas, USA
| | - Valerie Peterson
- Department of Thoracic Medical Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Manish A Shah
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Dung T Le
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
52
|
Vendrely V, Lemanski C, Pommier P, LE Malicot K, Saint A, Rivin Del Campo E, Regnault P, Baba-Hamed N, Ronchin P, Crehange G, Tougeron D, Menager-Tabourel E, Diaz O, Hummelsberger M, Minsat M, Drouet F, Larrouy A, Peiffert D, Lievre A, Zasadny X, Hautefeuille V, Mornex F, Lepage C, Quero L. Treatment, outcome, and prognostic factors in non-metastatic anal cancer: The French nationwide cohort study FFCD-ANABASE. Radiother Oncol 2023; 183:109542. [PMID: 36813175 DOI: 10.1016/j.radonc.2023.109542] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/24/2023]
Abstract
INTRODUCTION International guidelines regarding the treatment of squamous cell carcinoma of the anus (SCCA) recommend intensity-modulated radiotherapy (IMRT) combined with mitomycin-based chemotherapy (CT). The French FFCD-ANABASE cohort aimed at evaluating clinical practices, treatment, and outcomes of SCCA patients. METHODS This prospective multicentric observational cohort included all non-metastatic SCCA patients treated in 60 French centers from January 2015 to April 2020. Patients and treatment characteristics, colostomy-free survival (CFS), disease-free survival (DFS), overall survival (OS), and prognostic factors were analyzed. RESULTS Among 1015 patients (male: 24.4 %; female: 75.6 %; median age: 65 years), 43.3 %presented with early-stage(T1-2, N0) and 56.7 % with locally advanced stage (T3-4 or N + ) tumors. IMRT was used for 815 patients (80.3 %) and a concurrent CT was administered in 781 patients, consisting of mitomycin-based CT for 80 %. The median follow-up was 35.5 months. DFS, CFS, and OS at 3 years were 84.3 %, 85.6 %, and 91.7 % respectively in the early-stage group compared to 64.4 %, 66.9 %, and 78.2 % in the locally-advanced group (p < 0.001). In multivariate analyses, male gender, locally-advanced stage, and ECOG PS ≥ 1 were associated with poorer DFS, CFS, and OS. IMRT was significantly associated with a better CFS in the whole cohort and almost reached significance in the locally-advanced group. CONCLUSION Treatment of SCCA patients showed good respect for current guidelines. Significant differences in outcomes advocate for personalized strategies by either de-escalation for early-stage tumors or treatment intensification for locally-advanced tumors.
Collapse
Affiliation(s)
- Véronique Vendrely
- Department of Radiation Oncology, CHU Bordeaux, Bordeaux, France; BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, F-33000, Bordeaux, France.
| | - Claire Lemanski
- Department of Radiation Oncology, Montpellier Cancer Institute (ICM), Montpellier, France
| | | | - Karine LE Malicot
- Fédération Francophone de Cancérologie Digestive, university of Burgundy, Biostatistics, Dijon, France; EPICAD INSERM LNC-UMR 1231, University of Burgundy, Dijon, France
| | - Angélique Saint
- Department of Radiation Oncology, Antoine Lacassagne Cancer Center, Oncology, Nice, France
| | - Eleonor Rivin Del Campo
- Department of Radiation Oncology, Tenon University Hospital, APHP, Sorbonne University, Paris, France
| | | | | | | | - Gilles Crehange
- Radiotherapy department, Georges François Leclerc cancer center, Dijon, France
| | - David Tougeron
- Hepatology and Gastroenterology department, Poitiers University hospital, Poitiers, France
| | | | - Olivia Diaz
- Radiotherapy department, Daniel Hollard Institute, Grenoble, France
| | | | | | | | - Anne Larrouy
- Médical Oncology, Cancer institute, North Paris, France
| | - Didier Peiffert
- Department of radiaton oncology, Lorraine cancer center, Vandoeuvre-Les-Nancy, France
| | - Astrid Lievre
- Gastroenterology Department, Rennes University Hospital, Rennes 1 University, Inserm U1242 COSS (Chemistry Oncogenesis Stress Signaling, Rennes, France
| | - Xavier Zasadny
- Oncology radiotherapy department, Limoges polyclinic François Chenieux, Limoges, France
| | | | | | - Côme Lepage
- Department of hepato-gastroenterology, University hospital of Dijon, Dijon, France
| | - Laurent Quero
- INSERM U1160, Université Paris Cité, Paris, France; Radiotherapy Saint Louis Hospital, APHP, Paris, France
| |
Collapse
|
53
|
Sun L, Meng C, Zhang X, Gao J, Wei P, Zhang J, Zhang Z. Management and prediction of immune-related adverse events for PD1/PDL-1 immunotherapy in colorectal cancer. Front Pharmacol 2023; 14:1167670. [PMID: 37188271 PMCID: PMC10176603 DOI: 10.3389/fphar.2023.1167670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Programmed cell death protein (PD-1) is an important immunosuppressive molecule, which can inhibit interaction between PD-1 and its ligand PD-L1, further enhancing the T cell response and anti-tumor activity, which is called immune checkpoint blockade. Immunotherapy, represented by immune checkpoint inhibitors, has opened up a new era of tumor treatment and is gradually being applied to colorectal cancer recently. Immunotherapy was reported could achieve a high objective response rate (ORR) for colorectal cancer with high microsatellite instability (MSI), thus opening up a new era of colorectal cancer immunotherapy. Along with the increasing use of PD1 drugs in colorectal cancer, we should pay more attention to the adverse effects of these immune drugs while seeing the hope. Immune-related adverse events (irAEs) caused by immune activation and immune homeostasis during anti-PD-1/PD-L1 therapy can affect multi-organ and even be fatal in serious cases. Therefore, understanding irAEs is essential for their early detection and appropriate management. In this article, we review the irAEs that occur during the treatment of colorectal cancer patients with PD-1/PD-L1 drugs, analyze the current controversies and challenges, and point out future directions that should be explored, including exploring efficacy predictive markers and optimizing the paradigm of individualized immunotherapy.
Collapse
Affiliation(s)
- Liting Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Cong Meng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jiale Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Pengyu Wei
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jie Zhang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
54
|
Dhawan N, Afzal MZ, Amin M. Immunotherapy in Anal Cancer. Curr Oncol 2023; 30:4538-4550. [PMID: 37232801 DOI: 10.3390/curroncol30050343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
The incidence and mortality of squamous cell carcinoma of the anus has been gradually increasing globally over the last few decades. The evolution of different modalities, including immunotherapies, has changed the treatment paradigm of metastatic anal cancers. Chemotherapy, radiation therapy, and immune-modulating therapies form the backbone of treatment of anal cancer in various stages. Most anal cancers are linked to high-risk human papilloma virus (HPV) infections. HPV oncoproteins E6 and E7 are responsible for an anti-tumor immune response triggering the recruitment of tumor-infiltrating lymphocytes. This has led to the development and utilization of immunotherapy in anal cancers. Current research in anal cancer is moving forward to discover ways to incorporate immunotherapy in the treatment sequencing in various stages of anal cancers. Immune checkpoint inhibitors alone or in combination, adoptive cell therapy, and vaccines are the areas of active investigations in anal cancer in both locally advanced and metastatic settings. Immunomodulating properties of non-immunotherapies are incorporated to enhance immune checkpoint inhibitors' effectiveness in some of the clinical trials. The aim of this review is to summarize the potential role of immunotherapy in anal squamous cell cancers and future directions.
Collapse
Affiliation(s)
- Natasha Dhawan
- Department of Medical Oncology, Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Muhammad Z Afzal
- Department of Medical Oncology, Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Manik Amin
- Department of Medical Oncology, Dartmouth Cancer Center, Lebanon, NH 03756, USA
| |
Collapse
|
55
|
Morris VK, Jazaeri A, Westin SN, Pettaway C, George S, Huey RW, Grinsfelder M, Shafer A, Johnson B, Vining D, Guo M, Fellman B, Frumovitz M. Phase II Trial of MEDI0457 and Durvalumab for Patients With Recurrent/Metastatic Human Papillomavirus-Associated Cancers. Oncologist 2023:7146114. [PMID: 37104874 DOI: 10.1093/oncolo/oyad085] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 03/09/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Human papillomavirus (HPV) types 16/18 drive oncogenesis for most patients with cervical, anal, and penile cancers. MEDI0457, a therapeutic DNA vaccine containing plasmids for E6 and E7 HPV-16/18 viral oncogenes and IL-12 adjuvant, is safe and provokes an immune response against E6/E7. We tested MEDI0457 with the anti-PD-L1 antibody durvalumab for patients with HPV-associated cancers. METHODS Patients with recurrent/metastatic, treatment-refractory HPV-16/18 cervical cancer, or rare HPV-associated (anal and penile) cancers were eligible. Prior immune checkpoint inhibition was not permitted. Patients received MEDI0457 7 mg intramuscularly (weeks 1, 3, 7, 12, and every 8 weeks thereafter) and durvalumab 1500 mg intravenously every 4 weeks. The primary endpoint was overall response (RECIST 1.1). In this Simon two-stage phase 2 trial (Ho: p < 0.15; Ha: p ≥ 0.35), ≥2 responses were needed in both cervical and non-cervical cohorts during the first stage for the trial to proceed to stage 2 with an additional 25 patients (34 total) enrolled. RESULTS Twenty-one patients (12 cervical, 7 anal, and 2 penile) were evaluable for toxicity and 19 for response Overall response rate was 21% (95% CI, 6%-46%) among evaluable patients. Disease control rate was 37% (95% CI, 16%-62%). Median duration of response among responders was 21.8 months (95% CI, 9.7%-not estimable). Median progression-free survival was 4.6 months (95% CI, 2.8%-7.2%). Median overall survival was 17.7 months (95% CI, 7.6%-not estimable). Grades 3-4 treatment-related adverse events occurred in 6 (23%) participants. CONCLUSIONS The combination of MEDI0457 and durvalumab demonstrated acceptable safety and tolerability in patients with advanced HPV-16/18 cancers. The low ORR among patients with cervical cancer led to study discontinuation despite a clinically meaningful disease control rate.
Collapse
Affiliation(s)
- Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Amir Jazaeri
- Department of Gynecologic Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Curtis Pettaway
- Department of Urology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Solly George
- Department of Gynecologic Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Ryan W Huey
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Michaela Grinsfelder
- Department of Gynecologic Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Aaron Shafer
- Department of Gynecologic Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Benny Johnson
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - David Vining
- Department of Radiology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Guo
- Department of Pathology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Fellman
- Department of Biostatistics, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Frumovitz
- Department of Gynecologic Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
56
|
Monroy D, Serrano A, Galor A, Karp CL. Medical treatment for ocular surface squamous neoplasia. Eye (Lond) 2023; 37:885-893. [PMID: 36754986 PMCID: PMC10050251 DOI: 10.1038/s41433-023-02434-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 12/17/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Ocular surface squamous neoplasia (OSSN) is the most common non-melanocytic tumour of the ocular surface. Surgical excision with wide margins using the "no-touch" method was originally the most popular treatment for OSSN. However, in the past two decades, the use of topical medications for OSSN treatment has gained a reputation amongst ophthalmologists for being an effective alternative to surgical excision. Furthermore, technological advancements, such as those seen in high-resolution optical coherence tomography (HR-OCT) for the anterior segment, have facilitated the diagnosis and monitoring of OSSN. When selecting a topical agent, interferon alpha-2b (IFNα-2b) and 5-fluorouracil (5-FU) are two of the gentlest medications used for OSSN and are often considered first line therapies due to their high-resolution rates and mild side effect profiles. Mitomycin C (MMC), on the other hand, has a highly toxic profile; therefore, while effective, in our hands it is considered as a second-line treatment for OSSN if the other modalities fail. In addition, newer and less studied agents, such as immune checkpoint inhibitors, retinoic acid, aloe vera, and anti-vascular endothelial growth factor have anti-neoplastic properties and have shown potential for the treatment of OSSN. We enclose an updated literature review of medical treatments for OSSN.
Collapse
Grants
- R01 EY026174 NEI NIH HHS
- P30 EY014801 NEI NIH HHS
- I01 BX004893 BLRD VA
- R61 EY032468 NEI NIH HHS
- I01 CX002015 CSRD VA
- The Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development, Clinical Sciences R&D (CSRD) I01 CX002015 (Dr. Galor) and Biomedical Laboratory R&D (BLRD) Service I01 BX004893 (Dr. Galor), Department of Defense Gulf War Illness Research Program (GWIRP) W81XWH-20-1-0579 (Dr. Galor) and Vision Research Program (VRP) W81XWH-20-1-0820 (Dr. Galor), National Eye Institute R01EY026174 (Dr. Galor) and R61EY032468 (Dr. Galor), and Research to Prevent Blindness Unrestricted Grant (institutional).
- NIH Center Core Grant P30EY014801, RPB Unrestricted Award, Dr. Ronald and Alicia Lepke Grant, The Lee and Claire Hager Grant, The Robert Farr Family Grant, The Grant and Diana Stanton-Thornbrough Grant,The Robert Baer Family Grant, The Roberto and Antonia Menendez Grant, The Emilyn Page and Mark Feldberg Grant, The Calvin and Flavia Oak Support Fund, The Robert Farr Family Grant, The Jose Ferreira de Melo Grant, The Richard and Kathy Lesser Grant, The Honorable A. Jay Cristol Grant, The Michele and Ted Kaplan Grant, The Carol Soffer Grant, and the Richard Azar Family Grant(institutional grants).
Collapse
Affiliation(s)
- David Monroy
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andres Serrano
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Medical Center, New York, NY, USA
| | - Anat Galor
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Ophthalmology, Miami Veterans Administration Medical Center, Miami, FL, USA
| | - Carol L Karp
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
57
|
Azzi G, Tavallai M, Aushev VN, Koyen Malashevich A, Botta GP, Tejani MA, Hanna D, Krinshpun S, Malhotra M, Jurdi A, Aleshin A, Kasi PM. Using Tumor-Informed Circulating Tumor DNA (ctDNA)-Based Testing for Patients with Anal Squamous Cell Carcinoma. Oncologist 2023; 28:220-229. [PMID: 36562592 PMCID: PMC10020810 DOI: 10.1093/oncolo/oyac249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/27/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Anal squamous cell carcinoma (SCCA) is an uncommon malignancy with a rising incidence that has a high cure rate in its early stages. There is an unmet need for a reliable method to monitor response to treatment and assist in surveillance. Circulating tumor DNA (ctDNA) testing has shown great promise in other solid tumors for monitoring disease progression and detecting relapse in real time. This study aimed to determine the feasibility and use of personalized and tumor-informed ctDNA testing in SCCA. PATIENTS AND METHODS We analyzed real-world data from 251 patients (817 plasma samples) with stages I-IV SCCA, collected between 11/5/19 and 5/31/22. The tumor genomic landscape and feasibility of ctDNA testing was examined for all patients. The prognostic value of longitudinal ctDNA testing was assessed in patients with clinical follow-up (N = 37). RESULTS Whole-exome sequencing analysis revealed PIK3CA as the most commonly mutated gene, and no associations between mutations and stage. Anytime ctDNA positivity and higher ctDNA levels (MTM/mL) were associated with metastatic disease (P = .004). For 37 patients with clinical follow-up, median follow-up time was 21.0 months (range: 4.1-67.3) post-diagnosis. For patients with stages I-III disease, anytime ctDNA-positivity after definitive treatment was associated with reduced DFS (HR: 28.0; P = .005). CONCLUSIONS Our study demonstrates the feasibility of personalized and tumor-informed ctDNA testing as an adjunctive tool in patients with SCCA as well as potential use for detection of molecular/minuteimal residual disease, and relapse during surveillance. Prospective studies are needed to better evaluate the use of ctDNA testing in this indication.
Collapse
Affiliation(s)
- Georges Azzi
- HolyCross Medical Group, Ft. Lauderdale, FL, USA
| | | | | | | | | | | | - Diana Hanna
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | | | | | | | - Pashtoon M Kasi
- Corresponding author: Pashtoon M. Kasi, MD, MS, Department of Medical Oncology and Hematology, Weill Cornell Medicine, Englander Institute of Precision Medicine, Meyer Cancer Center, NewYork-Presbyterian Hospital, New York, NY 10021, USA. E-mail:
| |
Collapse
|
58
|
Anal Cancer: The Past, Present and Future. Curr Oncol 2023; 30:3232-3250. [PMID: 36975459 PMCID: PMC10047250 DOI: 10.3390/curroncol30030246] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Anal cancer is a rare cancer that accounts for about 2% of all gastrointestinal tract malignancies. Among anal cancer, squamous cell cancer is the most common malignancy. The incidence of all stages of anal squamous cell cancer has been increasing. Human papillomavirus infection and immunosuppression are major risk factors for anal cancer. The management of anal cancer has evolved over the past several decades and continues to do so. Chemoradiation therapy remains the mainstay for treatment for most patients with early-stage disease, whereas systemic therapy is the primary treatment for patients with metastatic disease. Patients with persistent disease or recurrence following chemoradiation therapy are treated with salvage surgery. Access to novel cytotoxic combinations and immunotherapy has improved the outcomes of patients with advanced disease. This review provides an overview of advances in the management of anal cancer over the past two decades. This paper reviews the epidemiology, risk factors, pathology, diagnosis, and management of localized and advanced anal squamous cell cancer, highlights current knowledge gaps in the management of anal cancer, and discusses future directions.
Collapse
|
59
|
Hernandez S, Das P, Holliday EB, Shen L, Lu W, Johnson B, Messick CA, Taniguchi CM, Skibber J, Ludmir EB, You YN, Smith GL, Bednarski B, Kostousov L, Koay EJ, Minsky BD, Tillman M, Portier S, Eng C, Koong AC, Chang GJ, Foo WC, Wang J, Soto LS, Morris VK. Differential Spatial Gene and Protein Expression Associated with Recurrence Following Chemoradiation for Localized Anal Squamous Cell Cancer. Cancers (Basel) 2023; 15:1701. [PMID: 36980587 PMCID: PMC10046657 DOI: 10.3390/cancers15061701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
The identification of transcriptomic and protein biomarkers prognosticating recurrence risk after chemoradiation of localized squamous cell carcinoma of the anus (SCCA) has been limited by a lack of available fresh tissue at initial presentation. We analyzed archival FFPE SCCA specimens from pretreatment biopsies prior to chemoradiation for protein and RNA biomarkers from patients with localized SCCA who recurred (N = 23) and who did not recur (N = 25). Tumor cells and the tumor microenvironment (TME) were analyzed separately to identify biomarkers with significantly different expression between the recurrent and non-recurrent groups. Recurrent patients had higher mean protein expression of FoxP3, MAPK-activation markers (BRAF, p38-MAPK) and PI3K/Akt activation (phospho-Akt) within the tumor regions. The TME was characterized by the higher protein expression of immune checkpoint biomarkers such as PD-1, OX40L and LAG3. For patients with recurrent SCCA, the higher mean protein expression of fibronectin was observed in the tumor and TME compartments. No significant differences in RNA expression were observed. The higher baseline expression of immune checkpoint biomarkers, together with markers of MAPK and PI3K/Akt signaling, are associated with recurrence following chemoradiation for patients with localized SCCA. These data provide a rationale towards the application of immune-based therapeutic strategies to improve curative-intent outcomes beyond conventional therapies for patients with SCCA.
Collapse
Affiliation(s)
- Sharia Hernandez
- Translational Molecular Pathology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA; (S.H.)
| | - Prajnan Das
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emma B. Holliday
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Li Shen
- Bioinformatics, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Lu
- Translational Molecular Pathology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA; (S.H.)
| | - Benny Johnson
- Gastrointestinal Medical Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Craig A. Messick
- Colon and Rectal Surgery, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cullen M. Taniguchi
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John Skibber
- Colon and Rectal Surgery, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ethan B. Ludmir
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y. Nancy You
- Colon and Rectal Surgery, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Grace Li Smith
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Brian Bednarski
- Colon and Rectal Surgery, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Larisa Kostousov
- Translational Molecular Pathology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA; (S.H.)
| | - Eugene J. Koay
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bruce D. Minsky
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew Tillman
- Colon and Rectal Surgery, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaelynn Portier
- Gastrointestinal Medical Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cathy Eng
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| | - Albert C. Koong
- Gastrointestinal Radiation Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George J. Chang
- Colon and Rectal Surgery, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wai Chin Foo
- Pathology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jing Wang
- Bioinformatics, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Luisa Solis Soto
- Translational Molecular Pathology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA; (S.H.)
| | - Van K. Morris
- Gastrointestinal Medical Oncology, The University of Texas—MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
60
|
Ito T, Takayanagi D, Sekine S, Hashimoto T, Shimada Y, Matsuda M, Yamada M, Hamamoto R, Kato T, Shida D, Kanemitsu Y, Boku N, Kohno T, Takashima A, Shiraishi K. Comparison of clinicopathological and genomic profiles in anal squamous cell carcinoma between Japanese and Caucasian cohorts. Sci Rep 2023; 13:3587. [PMID: 36869079 PMCID: PMC9984524 DOI: 10.1038/s41598-023-30624-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Anal squamous cell carcinoma (ASCC) is a rare tumor of the gastrointestinal tract. We aimed to compare the genetic backgrounds and their effect on clinical outcomes between Japanese and Caucasian patients with ASCC. Forty-one patients diagnosed with ASCC at the National Cancer Center Hospital were enrolled and evaluated for clinicopathological features, human papillomavirus (HPV) infection, HPV genotypes, p16 expression, PD-L1, and association of p16 status with the efficacy of concurrent chemoradiotherapy (CCRT). Target sequencing for hotspot mutations in 50 cancer-related genes was performed using genomic DNA from 30 available samples. Of 41 patients, 34 were HPV-positive (among them, HPV 16 was predominant; 73.2%); 38 patients were p16-positive (92.7%); and 39 patients received CCRT, of whom 36 were p16-positive and three p16-negative. p16-positive patients showed better complete response than p16-negative patients. Among 28 samples, 15 showed mutations in PIK3CA, FBXW7, ABL1, TP53, and PTEN; no difference in mutation profiles between the Japanese and Caucasian cohorts was observed. Actionable mutations were detected in both Japanese and Caucasian patients with ASCC. Genetic backgrounds, such as the HPV 16 genotype and PIK3CA mutations, were common regardless of ethnicity. p16 status may be a prognostic biomarker for CCRT in Japanese patients with ASCC.
Collapse
Affiliation(s)
- Takahiko Ito
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Daisuke Takayanagi
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigeki Sekine
- Department of Clinical Pathology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Taiki Hashimoto
- Department of Clinical Pathology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yoko Shimada
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Maiko Matsuda
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Masayoshi Yamada
- Endoscopy Division, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ryuji Hamamoto
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- RIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Dai Shida
- Department of Colorectal Surgery, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Division of Frontier Surgery, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Yukihide Kanemitsu
- Department of Colorectal Surgery, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Atsuo Takashima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
61
|
HPV virus and biomarkers of resistance to chemoradiation in circulating tumor cells from patients with squamous cell carcinoma of the anus. Pathol Res Pract 2023; 243:154327. [PMID: 36731178 DOI: 10.1016/j.prp.2023.154327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Localized anal cancer is mostly represented by squamous cell carcinoma of the anus (SCCA) and is cured in ≥80 % of cases by chemoradiation (CRT). Development of techniques for detection/evaluating circulating tumor cells (CTCs) for diagnosis/ prognosis/response to therapy can change the manner we treat/follow SCCA patients. OBJECTIVE to detect CTCs from patients with SCCA and evaluate the presence of HPV virus, p16 expression and markers related to resistance to CRT (RAD23B/ ERCC1/ TYMS) in CTCs at baseline and after CRT. METHODS CTCs were isolated/quantified by ISET®, protein expressions were analyzed by immunocytochemistry and HPV DNA was detected by chromogenic in situ hybridization. RESULTS We enrolled 15 patients: median age was 61 (43-73) years, the majority was women (10/15). CTCs were detected in all patients at baseline (median= 0.4 (0.4-3.33) CTCs/mL) and in 8/9 patients, after CRT (median= 2.33 (0-7.0) CTCs/mL). DNA from HPV was found in CTCs in 14/15 patients (93.33 %) at baseline and in 7/9 (77.7 %) after treatment. At a median follow-up of 22.20 (1.45-38.55) months, three patients expressed ERCC1 in CTCs after treatment, with one of them having disease recurrence. CONCLUSION We showed that detection of HPV in CTCs from patients with non-metastatic SCCA is feasible and appears to be a sensitive diagnostic method. These results may be clinically useful for better monitoring these patients. However, future larger cohorts may demonstrate whether there is any correlation between the presence of HPV and the expression of screening markers for CRT in SCCA.
Collapse
|
62
|
Prete AA, Manca P, Messina M, Formica V, Frassineti GL, Zampino MG, Corsi DC, Orciuolo C, Prisciandaro M, Bergamo F, Angerilli V, Scartozzi M, Casagrande M, Masi G, Ronzoni M, Morano F, Vettore V, Salmaso R, Rasola C, Maddalena G, Del Bianco P, Milione M, Cremolini C, Fassan M, Pietrantonio F, Lonardi S. Extensive molecular profiling of squamous cell anal carcinoma in a phase 2 trial population: Translational analyses of the "CARACAS" study. Eur J Cancer 2023; 182:87-97. [PMID: 36753836 DOI: 10.1016/j.ejca.2022.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/02/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Molecular characteristics of squamous cell anal carcinoma (SCAC) are poorly explored. Immune checkpoint inhibitors showed limited activity in phase I/II trials, but predictive and prognostic biomarkers are lacking. PATIENTS AND METHODS In the phase II randomised trial CARACAS (NCT03944252), avelumab alone (Arm A) or with cetuximab (Arm B) was tested in pre-treated advanced SCAC , with overall response rate being the primary end-point. On pre-treatment tumour tissue samples, we assessed Human papillomavirus status, programmed-death ligand 1 (PD-L1) expression, mismatch repair proteins expression, tumour mutational burden (TMB) and comprehensive genomic profiling by FoundationOne CDx. Tumour-infiltrating lymphocytes were characterised on haematoxylin-eosine-stained samples. Primary objective was to describe response to immunotherapy in the CARACAS trial population according to molecular and histological characteristics. Secondary objectives were to assess progression-free survival (PFS) and overall survival (OS) according to molecular biomarkers. RESULTS High PD-L1 (>40 with combined positive score) was significantly more frequent in patients with disease control (p = 0.0109). High TMB (>10 mutations per megabase) was related to better OS (hazard ratio (HR) = 0.09; 95%confidence interval (CI) 0.01-0.68; p = 0.019) and PFS (HR = 0.44; 95%CI = 0.15-1.27; p = 0.129). High expression of PD-L1 conferred longer OS (HR = 0.46; 95%CI = 0.19-1.08; p = 0.075) and PFS (HR = 0.42; 95%CI = 0.20-0.92; p = 0.03). Neither OS (HR = 1.30; 95%CI = 0.72-2.36; p = 0.39) or PFS (HR = 1.31; 95%CI = 0.74-2.31; p = 0.357) was affected by high (>1.2) Tumour-infiltrating lymphocytes count. High TMB and PD-L1identified patients were with significantly better OS (HR = 0.33; 95%CI = 0.13-0.81; p = 0.015) and PFS (HR = 0.48; 95%CI = 0.23-1.00; p = 0.015). CONCLUSIONS To our knowledge, TranslaCARACAS is the first study to document prognostic role of TMB and PD-L1 in advanced SCAC patients treated with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Alessandra A Prete
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Paolo Manca
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy
| | - Marco Messina
- Oncologia, Fondazione Istituto G. Giglio, Cefalù, Italy
| | | | - Giovanni L Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumouri "Dino Amadori" (IRST), Meldola, Italy
| | - Maria G Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumours, European Institute of Oncology - IRCCS, Milan, Italy
| | - Domenico C Corsi
- Medical Oncology Unit Ospedale San Giovanni Calibita Fatebenefratelli, Rome, Italy
| | - Corrado Orciuolo
- Oncology Unit, Department of Radiology, Oncology and Human Pathology, Sapienza University of Rome, Italy
| | | | - Francesca Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua Padua, Italy
| | | | | | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Monica Ronzoni
- Oncologia Medica, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Morano
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy
| | - Valentina Vettore
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Roberta Salmaso
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua Padua, Italy
| | - Cosimo Rasola
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy
| | - Giulia Maddalena
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Massimo Milione
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumouri, Milan, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy.
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua Padua, Italy; Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Sara Lonardi
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
63
|
Efficacy and tolerance of cetuximab in combination with 5 FU plus irinotecan based chemotherapy in metastatic squamous cell anal carcinoma. Dig Liver Dis 2023; 55:407-411. [PMID: 36088220 DOI: 10.1016/j.dld.2022.08.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Squamous cell anal carcinoma (SCAC) is an uncommon neoplasia often cured by surgery and/or chemo-adiation therapy at the localized stage. Although the first-line treatment for metastatic anal canal cancer is now better codified with two validated treatment regimens, carboplatin-paclitaxel and modified docetaxel-cisplatin-5FU (DCF), there is little data and no consensus regarding subsequent lines [1-5]. In this study, we report the safety and efficacy of cetuximab (an epidermal growth factor receptor inhibitor) in combination with 5-FU plus irinotecan based chemotherapy. METHOD A retrospective analysis of patients with metastatic SCAC (mSCAC), who failed on at least one prior line of treatment, before being treated with the combination FOLFIRI and cetuximab between March 2015 and February 2022 at Gustave Roussy cancer center, was performed. RESULTS A total of 33 patients with a pre-treated mSCAC were analyzed. The combination of FOLFIRI and cetuximab provided a disease control rate (DCR) of 73%, and response rate of 30%. With a median follow-up of 38 months, the median progression free survival was 5.5 months, and the median overall survival was 13.7 months. Fourteen patients (42%) experienced grade III/IV adverse events that remained manageable. CONCLUSION Our study suggests that FOLFIRI and cetuximab is a promising combination in the management of mSCAC with a very good DCR and a manageable toxicity profile. Further prospective trials would be needed to confirm our results.
Collapse
|
64
|
Li Y, Shen F, Tan Q, Chen Y, Gu Y. Research Progress of Immuno-Inhibitory Receptors in Gynecological Cervical Cancer. Technol Cancer Res Treat 2023; 22:15330338231208846. [PMID: 37908109 PMCID: PMC10621300 DOI: 10.1177/15330338231208846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/03/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
The mortality rate of cervical cancer is the highest among female malignant tumors and seriously threatens women's lives and health. Persistent high-risk human papillomavirus (HPV) infection is the leading cause of cervical cancer, which provides the basis for immunotherapy. In recent years, owing to progress in targeted therapy and immunotherapy, the survival time of patients with cervical cancer has been significantly extended. However, effective treatments for advanced, recurrent, and metastatic cancers are lacking. "Tumor immunotherapy" has been described as a viable option for tumor therapy but the efficacy of immunotherapy for cervical cancer has only been demonstrated in phase I or II clinical trials. Immune checkpoint inhibitors (ICIs) have shown promising clinical results particularly for treating recurrent and advanced cervical cancer, however, they remain inadequate in some patients. Immune checkpoint is the target of immunotherapy. Therefore, the identification of novel therapeutic targets is essential. In this paper, the structure, expression, function, biological effect of immune inhibitory receptors (IRs) and related clinical studies were reviewed, in order to further explore the application potential of these immune checkpoints and apply them to the future clinical treatment of cervical cancer.
Collapse
Affiliation(s)
- Yang Li
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Gynecology and obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fangrong Shen
- Department of Gynecology and obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qingqing Tan
- Department of Gynecological Oncology, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Youguo Chen
- Department of Gynecology and obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
65
|
Pourhamzeh M, Asadian S, Mirzaei H, Minaei A, Shahriari E, Shpichka A, Es HA, Timashev P, Hassan M, Vosough M. Novel antigens for targeted radioimmunotherapy in hepatocellular carcinoma. Mol Cell Biochem 2023; 478:23-37. [PMID: 35708866 DOI: 10.1007/s11010-022-04483-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/18/2022] [Indexed: 01/17/2023]
Abstract
Liver cancer is the sixth common cancer and forth cause of cancer-related death worldwide. Based on usually advanced stages of hepatocellular carcinoma (HCC) at the time of diagnosis, therapeutic options are limited and, in many cases, not effective, and typically result in the tumor recurrence with a poor prognosis. Radioimmunotherapy (RIT) offers a selective internal radiation therapy approach using beta or alpha emitting radionuclides conjugated with tumor-specific monoclonal antibodies (mAbs), or specific selective peptides. When compared to chemotherapy or radiotherapy, radiolabeled mAbs against cancer-associated antigens could provide a high therapeutic and exclusive radiation dose for cancerous cells while decreasing the exposure-induced side effects to healthy tissues. The recent advances in cancer immunotherapy, such as blockade of immune-checkpoint inhibitors (ICIs), has changed the landscape of cancer therapy, and the efficacy of different classes of immunotherapy has been tested in many clinical trials. Taking into account the use of ICIs in the liver tumor microenvironment, combined therapies with different approaches may enhance the outcome in the future clinical studies. With the development of novel immunotherapy treatment options in the recent years, there has been a great deal of information about combining the diverse treatment modalities to boost the effectiveness of immunomodulatory drugs. In this opinion review, we will discuss the recent advancements in RIT. The current status of immunotherapy and internal radiotherapy will be updated, and we will propose novel approaches for the combination of both techniques. Potential target antigens for radioimmunotherapy in Hepatocellular carcinoma (HCC). HCC radioimmunotherapy target antigens are the most specific and commonly accessible antigens on the surface of HCC cells. CTLA-4 ligand and receptor, TAMs, PD-1/PD-L, TIM-3, specific IEXs/TEXs, ROBO1, and cluster of differentiation antigens CD105, CD147 could all be used in HCC radioimmunotherapy. Abbreviations: TAMs, tumor-associated macrophages; CTLA-4, cytotoxic T-lymphocyte associated antigen-4; PD-1, Programmed cell death protein 1; PD-L, programmed death-ligand1; TIM-3, T-cell immunoglobulin (Ig) and mucin-domain containing protein-3; IEXs, immune cell-derived exosomes; TEXs, tumor-derived exosomes.
Collapse
Affiliation(s)
- Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Departments of Pathology and Medicine, UC San Diego, La Jolla, CA, USA
| | - Samieh Asadian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Azita Minaei
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Elahe Shahriari
- Departments of Pathology and Medicine, UC San Diego, La Jolla, CA, USA
| | - Anastasia Shpichka
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | | | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia. .,Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia. .,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
66
|
Wang H, Xu Y, Zuo F, Liu J, Yang J. Immune-based combination therapy for esophageal cancer. Front Immunol 2022; 13:1020290. [PMID: 36591219 PMCID: PMC9797857 DOI: 10.3389/fimmu.2022.1020290] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Esophageal cancer (EC) is an aggressive malignancy raising a healthcare concern worldwide. Standard treatment options include surgical resection, chemotherapy, radiation therapy, and targeted molecular therapy. The five-year survival rate for all stages of EC is approximately 20%, ranging from 5% to 47%, with a high recurrence rate and poor prognosis after treatment. Immunotherapy has shown better efficacy and tolerance than conventional therapies for several malignancies. Immunotherapy of EC, including immune checkpoint inhibitors, cancer vaccines, and adoptive cell therapy, has shown clinical advantages. In particular, monoclonal antibodies against PD-1 have a satisfactory role in combination therapy and are recommended for first- or second-line treatments. Here, we present a systematic summary and analysis of immunotherapy-based combination therapies for EC.
Collapse
Affiliation(s)
- Huiling Wang
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yufei Xu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Fengli Zuo
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Junzhi Liu
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiqiao Yang
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China,Breast Center, West China Hospital of Sichuan University, Chengdu, China,*Correspondence: Jiqiao Yang,
| |
Collapse
|
67
|
The emerging role of immunotherapy in the treatment of anal cancer. Curr Opin Pharmacol 2022; 67:102309. [PMID: 36334330 DOI: 10.1016/j.coph.2022.102309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/19/2022] [Indexed: 01/25/2023]
Abstract
For decades, chemoradiotherapy for early-stage disease and systemic chemotherapy for advanced disease have represented the mainstay of treatment for anal cancer. Over the last few years, however, the advent of immunotherapy has opened interesting therapeutic perspectives, with the establishment of new standards of care, and the development of clinical trials that may further shape the treatment algorithm for this tumour. In this review article, we discuss the rationale behind the use of immunotherapy for anal cancer and provide an overview of the available clinical data and ongoing efforts to build on these.
Collapse
|
68
|
Hervé L, Kim S, Boustani J, Klajer E, Pernot M, Nguyen T, Lakkis Z, Borg C, Vienot A. Modified DCF (Docetaxel, Cisplatin and 5-fluorouracil) chemotherapy is effective for the treatment of advanced rectal squamous cell carcinoma. Front Oncol 2022; 12:974108. [DOI: 10.3389/fonc.2022.974108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
BackgroundAdvanced rectal squamous cell carcinoma (rSCC) is a very rare and aggressive entity, and the best initial management is crucial for long survival as well as organ preservation and quality of life. Whereas local diseases are treated with chemo-radiotherapy and salvage surgery, data are scarce on how to treat more advanced diseases, and the role of induction chemotherapy is unknown.MethodsWe retrospectively analyzed all consecutive patients with advanced rSCC and treated with modified DCF (docetaxel, cisplatin, 5-fluorouracil; mDCF) regimen, from January 2014 and December 2021 in two French centers. Exploratory endpoints were efficacy (overall survival, recurrence-free survival, response rate, organ preservation rate) and safety.ResultsNine patients with locally advanced or metastatic diseases received a mDCF regimen and were included for analysis. The median age was 62.0 years, 7 patients (77.8%) were women, and all eight available tumors were positive for HPV, mostly (85.7%) to genotype 16. With a median follow-up of 33.1 months, 77.8% of patients were still alive and disease-free, and the median overall survival was not reached at six years. The objective response rate was 87.5% after mDCF, and the complete response rate was 25.0% after mDCF and was increased to 75.0% after chemoradiotherapy. Only one patient underwent surgery on the primary tumor, with a complete pathological response. The median mDCF cycle was eight over eight scheduled, and all patients received the complete dose of radiotherapy without interruptions.ConclusionsInduction mDCF chemotherapy followed by chemoradiotherapy is safe and highly effective in patients with advanced rSCC, and should be considered as an option in metastatic stage or locally advanced disease with an organ-preservation strategy.
Collapse
|
69
|
Debernardi A, Meurisse A, Prétet JL, Guenat D, Monnien F, Spehner L, Vienot A, Roncarati P, André T, Abramowitz L, Molimard C, Mougin C, Herfs M, Kim S, Borg C. Prognostic role of HPV integration status and molecular profile in advanced anal carcinoma: An ancillary study to the epitopes-HPV02 trial. Front Oncol 2022; 12:941676. [PMID: 36313663 PMCID: PMC9614213 DOI: 10.3389/fonc.2022.941676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/20/2022] [Indexed: 02/25/2025] Open
Abstract
Squamous Cell Carcinoma of the Anal canal (SCCA) is a rare disease associated with a Human Papillomavirus (HPV) infection in most cases, predominantly the HPV16 genotype. About 15% of SCCA are diagnosed in metastatic stage and some will relapse after initial chemoradiotherapy (CRT). Treatment of patients by Docetaxel, Cisplatin and 5-fluorouracil (DCF) has been recently shown to improve their complete remission and progression-free survival. The aim of this retrospective study was to explore the impact of HPV infection, HPV DNA integration, TERT promoter mutational status and somatic mutations of oncogenes on both progression-free (PFS) and overall survivals (OS) of patients treated by DCF. Samples obtained from 49 patients included in the Epitopes-HPV02 clinical trial, diagnosed with metastatic or non-resectable local recurrent SCCA treated by DCF, were used for analyses. Median PFS and OS were not associated with HPV status. Patients with episomal HPV had an improved PFS compared with SCCA patients with integrated HPV genome (p=0.07). TERT promoter mutations were rarely observed and did not specifically distribute in a subset of SCCA and did not impact DCF efficacy. Among the 42 genes investigated, few gene alterations were observed, and were in majority amplifications (68.4%), but none were significantly correlated to PFS. As no biomarker is significantly associated with patients' survival, it prompts us to include every patient failing CRT or with metastatic disease in DCF strategy.
Collapse
Affiliation(s)
- Alice Debernardi
- EA3181, University of Bourgogne Franche-Comté, LabEx LipSTIC ANR-11-LABX-0021, Besançon, France
| | - Aurélia Meurisse
- Methodology and Quality of Life in Oncology Unit, University Hospital of Besançon, Besançon, France
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
| | - Jean-Luc Prétet
- EA3181, University of Bourgogne Franche-Comté, LabEx LipSTIC ANR-11-LABX-0021, Besançon, France
- Papillomavirus National Reference Center, University Hospital, Besançon, France
| | - David Guenat
- EA3181, University of Bourgogne Franche-Comté, LabEx LipSTIC ANR-11-LABX-0021, Besançon, France
- Molecular Biology and Microbiology Department, Anamed SA Laboratory, Lausanne, Switzerland
| | - Franck Monnien
- Department of Pathology, University Hospital of Besançon, Besançon, France
| | - Laurie Spehner
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Angélique Vienot
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Patrick Roncarati
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Thierry André
- Department of Medical Oncology, University Hospital Saint Antoine, Paris, France
| | - Laurent Abramowitz
- Division of Gastroenterology and Hepatology and Proctology, University Hospital Bichat, Paris, France
- Ramsay GDS, Blomet Clinic, Paris, France
| | - Chloé Molimard
- Department of Anatomopathology, University Hospital of Besançon, Besançon, France
| | - Christiane Mougin
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
- Papillomavirus National Reference Center, University Hospital, Besançon, France
| | - Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Stefano Kim
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
- Clinical investigation center, CIC-1403 University Hospital of Besançon, Besançon, France
- Department of Medical Oncology, Sanatorio Allende, Cordoba, Argentina
| | - Christophe Borg
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
- Molecular Biology and Microbiology Department, Anamed SA Laboratory, Lausanne, Switzerland
- Clinical investigation center, CIC-1403 University Hospital of Besançon, Besançon, France
| |
Collapse
|
70
|
Chan AMY, Roldan Urgoiti G, Jiang W, Lee S, Kornaga E, Mathen P, Yeung R, Enwere EK, Box A, Konno M, Koebel M, Joseph K, Doll CM. The prognostic impact of PD-L1 and CD8 expression in anal cancer patients treated with chemoradiotherapy. Front Oncol 2022; 12:1000263. [PMID: 36276142 PMCID: PMC9585228 DOI: 10.3389/fonc.2022.1000263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022] Open
Abstract
Background Programmed death-ligand 1 (PD-L1) expression has been shown to be prognostic in many cancer types and used in consideration of checkpoint inhibitor immunotherapy. However, there are very limited and conflicting data on the prognostic impact of PD-L1 in patients with anal squamous cell carcinoma (ASCC). The objectives of this study were to measure the expression of PD-L1 and CD8 in patients with ASCC treated with radical chemoradiotherapy (CRT) and to correlate tumor expression with progression-free survival (PFS) and overall survival (OS). Methods Ninety-nine patients with ASCC treated with primary CRT at two tertiary care cancer centers between 2000 and 2013, with available pre-treatment tumors, were included. Tissue microarrays (TMAs) from pre-treatment tumor specimens were stained for PD-L1 and CD8. PD-L1 expression in the tumor and stroma was quantified using HALO image analysis software, and results were interpreted using quantitative methods. The density of CD8 cells within the tumor was interpreted by a trained pathologist semi-quantitatively, using a 0-4 scoring system. Kaplan-Meier analysis with log-rank was used to determine the significance in the association of tumor markers with PFS and OS. Cox multivariate analysis was used to explore independent predictors of PFS and OS. Results Of the 99 patients, 63 (64%) had sufficient tumor samples available for full analysis. CD8 high status was documented in 32 of 63 (50.8%) % of cases. PD-L1 expression was positive in 88.9% of cases. Approximately half the patients had tumor PD-L1 ≥ 5%. Patients with tumor PD-L1 ≥ 5% had better OS vs those with lower expression, HR=0.32 (95% CI 0.11-0.87), p=0.027; 10 years OS: 84% for tumor PD-L1 ≥ 5% vs 49% for PD-L1 < 5%. PD-L1 expression was not associated with PFS. On multivariate analysis, tumor PD-L1 ≥ 5% showed a trend to statistical significance for better OS, HR=0.55 (95% CI 0.12- 1.00), p=0.052. Conclusions Tumor PD-L1≥5% is associated with OS in patients with ASCC treated with CRT. PD-L1 expression status using this unique cut-point warrants further validation for prognostication in patients with this disease. Future studies are required to determine the benefit of alternative treatment strategies based on PD-L1 status.
Collapse
Affiliation(s)
- Angela MY. Chan
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | | | - Will Jiang
- Division of Radiation Oncology, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Lee
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Elizabeth Kornaga
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Peter Mathen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Rosanna Yeung
- Department of Radiation Oncology, Evergreen Health, Kirkland, WA, United States
| | - Emeka K. Enwere
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Alan Box
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Regina, SK, Canada
| | - Mie Konno
- Precision Oncology Hub, Tom Baker Cancer Centre, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Martin Koebel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Kurian Joseph
- Division of Radiation Oncology, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Corinne M. Doll
- Department of Oncology, University of Calgary, Calgary, AB, Canada
- *Correspondence: Corinne M. Doll,
| |
Collapse
|
71
|
Mastracci L, Grillo F, Parente P, Gullo I, Campora M, Angerilli V, Rossi C, Sacramento ML, Pennelli G, Vanoli A, Fassan M. PD-L1 evaluation in the gastrointestinal tract: from biological rationale to its clinical application. Pathologica 2022; 114:352-364. [PMID: 36305021 PMCID: PMC9614301 DOI: 10.32074/1591-951x-803] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
Immune-checkpoint inhibitors targeting the PD-1/PD-L1 axis have brought significant clinical benefit in many solid cancer types, including gastrointestinal malignancies. However, it has been estimated that only 20-40% of patients respond to treatment. The pattern of expression and potential predictive value of PD-L1 as an immunohistochemical biomarker has been extensively studied in gastrointestinal neoplasms. Until now, its predictive value has been demonstrated, and is currently in use only in upper gastrointestinal malignancies (gastroesophageal adenocarcinoma and esophageal squamous cell carcinoma). In this Review, we describe the technical aspects and challenges related to PD-L1 immunohistochemical assays, the current role of PD-L1 as a biomarker in clinical practice and we outline the main studies and clinical trials analyzing the prognostic and predictive value of PD-L1 in gastrointestinal cancers.
Collapse
Affiliation(s)
- Luca Mastracci
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| | - Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Irene Gullo
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
- Department of Pathology, Faculty of Medicine of the University of Porto (FMUP), Portugal
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Portugal
| | - Michela Campora
- Public Healthcare Trust of the Autonomous Province of Trento, Santa Chiara Hospital, Department of Laboratory Medicine, Pathology Unit, Trento, Italy
| | - Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
| | - Chiara Rossi
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Pavia, Italy
| | - Maria Luisa Sacramento
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
| | - Gianmaria Pennelli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Pavia, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua (PD), Italy
| |
Collapse
|
72
|
Rogers JE, Yamashita K, Sewastjanow Silva M, Ajani JA. Current Immune Checkpoint Inhibitor Genetic Biomarker Exploration in Gastrointestinal Tumors. Cancers (Basel) 2022; 14:4804. [PMID: 36230726 PMCID: PMC9563283 DOI: 10.3390/cancers14194804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/16/2022] [Accepted: 09/24/2022] [Indexed: 11/19/2022] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer management. Some patients with gastrointestinal (GI) tract malignancy have experienced remarkable results. Here, in our review, we discuss predictive/prognostic GI tumor biomarkers that appear to correlate with benefits with this strategy. Remarkable progress has been made in certain subsets of patients including the potential for solid tumor patients to avoid local therapies such as radiation and/or surgery (organ preservation), which come with acute and chronic risks that have historically been the only curable strategies for these GI tumors. These results provide new and exciting strategies for solid tumor management. Unfortunately, immune checkpoint inhibitors can correlate with biomarkers, but benefits occur in a small subset of patients with GI malignancies. Most frequently, immune checkpoint inhibitors fail to induce response in GI malignancies due to the "cold" tumor microenvironment that protects cancer. Translational strategies are needed to develop effective combination strategies and novel biomarkers to overcome the intrinsic resistance.
Collapse
Affiliation(s)
- Jane E. Rogers
- U.T. M.D. Anderson Cancer Center Pharmacy Clinical Programs, Houston, TX 77030, USA
| | - Kohei Yamashita
- U.T. M.D. Anderson Cancer Center Department of Gastrointestinal Medical Oncology, Houston, TX 77030, USA
| | - Matheus Sewastjanow Silva
- U.T. M.D. Anderson Cancer Center Department of Gastrointestinal Medical Oncology, Houston, TX 77030, USA
| | - Jaffer A. Ajani
- U.T. M.D. Anderson Cancer Center Department of Gastrointestinal Medical Oncology, Houston, TX 77030, USA
| |
Collapse
|
73
|
Demes M, Pession U, Jeroch J, Schulze F, Eichler K, Martin D, Wild P, Waidmann O. Site of analysis matters - Ongoing complete response to Nivolumab in a patient with HIV/HPV related metastatic anal cancer and MLH1 mutation. Oncotarget 2022; 13:1034-1042. [PMID: 36128324 PMCID: PMC9477220 DOI: 10.18632/oncotarget.28274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 09/05/2022] [Indexed: 01/16/2023] Open
Abstract
Anal cancer is a rare disease with increasing incidence. In patients with locally recurrent or metastatic disease which cannot be treated with chemoradiotherapy or salvage surgery systemic first-line chemotherapy with carboplatin and paclitaxel is standard of care. For patients who progress after first-line therapy and are still eligible for second-line therapy Programmed cell death protein 1 (PD-1) antibodies are potential therapeutic options. However, prediction of response to immunotherapy is still challenging including anal cancer. We report here to our knowledge the first anal cancer case with microsatellite instability (MSI) due to MLH1 mutation and a deep and ongoing response to Nivolumab treatment. Namely, thorough analysis of the primary tumor as well as metastatic sites by next generation sequencing (NGS) revealed that MSI was formally only found in the metastatic sites but not in the primary tumor. Concomitantly, tumor mutational burden (TMB) was higher in the metastatic site than in the primary tumor. Therefore, we conclude that all anal cancer patients should be tested for MSI and whenever possible molecular analysis should be performed rather from metastatic sites than from the primary tumor.
Collapse
Affiliation(s)
- Melanie Demes
- Dr. Senckenbergisches Institut für Pathologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Ursula Pession
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Jan Jeroch
- Dr. Senckenbergisches Institut für Pathologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Falko Schulze
- Dr. Senckenbergisches Institut für Pathologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Katrin Eichler
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Daniel Martin
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Klinik für Strahlentherapie und Onkologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Peter Wild
- Dr. Senckenbergisches Institut für Pathologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
| | - Oliver Waidmann
- Universitäres Centrum für Tumorerkrankungen (UCT), Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Medizinische Klinik 1, Schwerpunkte Gastroenterologie und Hepatologie, Universitätsklinikum Frankfurt, Frankfurt 60590, Germany
- Centrum für Hämatologie und Onkologie, Frankfurt 60389, Germany
| |
Collapse
|
74
|
Kiran RP, Kochhar GS, Kariv R, Rex DK, Sugita A, Rubin DT, Navaneethan U, Hull TL, Ko HM, Liu X, Kachnic LA, Strong S, Iacucci M, Bemelman W, Fleshner P, Safyan RA, Kotze PG, D'Hoore A, Faiz O, Lo S, Ashburn JH, Spinelli A, Bernstein CN, Kane SV, Cross RK, Schairer J, McCormick JT, Farraye FA, Chang S, Scherl EJ, Schwartz DA, Bruining DH, Philpott J, Bentley-Hibbert S, Tarabar D, El-Hachem S, Sandborn WJ, Silverberg MS, Pardi DS, Church JM, Shen B. Management of pouch neoplasia: consensus guidelines from the International Ileal Pouch Consortium. Lancet Gastroenterol Hepatol 2022; 7:871-893. [PMID: 35798022 DOI: 10.1016/s2468-1253(22)00039-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
Surveillance pouchoscopy is recommended for patients with restorative proctocolectomy with ileal pouch-anal anastomosis in ulcerative colitis or familial adenomatous polyposis, with the surveillance interval depending on the risk of neoplasia. Neoplasia in patients with ileal pouches mainly have a glandular source and less often are of squamous cell origin. Various grades of neoplasia can occur in the prepouch ileum, pouch body, rectal cuff, anal transition zone, anus, or perianal skin. The main treatment modalities are endoscopic polypectomy, endoscopic ablation, endoscopic mucosal resection, endoscopic submucosal dissection, surgical local excision, surgical circumferential resection and re-anastomosis, and pouch excision. The choice of the treatment modality is determined by the grade, location, size, and features of neoplastic lesions, along with patients' risk of neoplasia and comorbidities, and local endoscopic and surgical expertise.
Collapse
Affiliation(s)
- Ravi P Kiran
- Division of Colorectal Surgery, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA
| | - Gursimran S Kochhar
- Division of Gastroenterology, Hepatology, and Nutrition, Allegheny Health Network, Pittsburgh, PA, USA
| | - Revital Kariv
- Department of Gastroenterology, Tel Aviv Sourasky Medical Center and Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Douglas K Rex
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Akira Sugita
- Department of Clinical Research and Department of inflammatory Bowel Disease, Yokohama Municipal Citizens Hospital Yokohama, Japan
| | - David T Rubin
- University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago, IL, USA
| | - Udayakumar Navaneethan
- IBD Center and IBD Interventional Unit, Center for Interventional Endoscopy, Orlando Health, Orlando, FL, USA
| | - Tracy L Hull
- Department of Colorectal Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Huaibin Mabel Ko
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA
| | - Xiuli Liu
- Department of Pathology and Immunology, Washington University, St Louis, MO, USA
| | - Lisa A Kachnic
- Department of Radiation Oncology, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA
| | - Scott Strong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marietta Iacucci
- Institute of Immunology and Immunotherapy, NIHR Birmingham Biomedical Research Centre, University Hospitals NHS Foundation Trust, University of Birmingham, UK
| | - Willem Bemelman
- Department of Surgery, Academic Medical Center, Amsterdam, Netherlands
| | - Philip Fleshner
- Division of Colorectal Surgery, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Rachael A Safyan
- Division of Hematology and Oncology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Paulo G Kotze
- IBD Outpatients Clinic, Catholic University of Paraná, Curitiba, Brazil
| | - André D'Hoore
- Department of Abdominal Surgery, University Hospital Leuven, Belgium
| | - Omar Faiz
- Department of Surgery, St Mark's Hospital and Academic Institute, Harrow and Department of Surgery and Cancer, Imperial College London, London, UK
| | - Simon Lo
- Pancreatic and Biliary Disease Program, Digestive Diseases, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Jean H Ashburn
- Department of Surgery, Wake Forest University Baptist Medical Center, Winston-Salem, NC, USA
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Division Colon and Rectal Surgery, Rozzano, Milan, Italy
| | - Charles N Bernstein
- University of Manitoba Inflammatory Bowel Disease Clinical and Research Centre, Winnipeg, MB, Canada
| | - Sunanda V Kane
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Raymond K Cross
- Inflammatory Bowel Disease Program, University of Maryland School of Medicine, MD, USA
| | - Jason Schairer
- Department of Gastroenterology, Henry Ford Health System, Detroit, MI, USA
| | - James T McCormick
- Division of Colon and Rectal Surgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Francis A Farraye
- Division of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Shannon Chang
- Inflammatory Bowel Disease Center, NYU Langone Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Ellen J Scherl
- Jill Roberts Center for IBD, Gastroenterology and Hepatology, Weill Cornell Medicine and NewYork Presbyterian Hospital, New York, NY, USA
| | - David A Schwartz
- Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David H Bruining
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Jessica Philpott
- Department of Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic, Cleveland, OH, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA
| | - Dino Tarabar
- IBD Clinical Center, University Hospital Center Dr Dragiša Mišović, Belgrade, Serbia
| | - Sandra El-Hachem
- Division of Gastroenterology, Hepatology, and Nutrition, Allegheny Health Network, Pittsburgh, PA, USA
| | - William J Sandborn
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Mark S Silverberg
- Mount Sinai Hospital Inflammatory Bowel Disease Centre, Toronto, ON, Canada
| | - Darrell S Pardi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - James M Church
- Division of Colorectal Surgery, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA
| | - Bo Shen
- Center for Interventional Inflammatory Bowel Disease, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
75
|
Immune Checkpoint Inhibitors for Gastrointestinal Malignancies: An Update. Cancers (Basel) 2022; 14:cancers14174201. [PMID: 36077740 PMCID: PMC9454768 DOI: 10.3390/cancers14174201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Immune checkpoint inhibitors are a class of anti-cancer therapy that work by harnessing the body’s immune system to promote cancer cell death. These drugs have become standard of care for many types of cancer, including melanoma and lung cancer, after clinical trials showed they work better than traditional chemotherapy. The role of immune checkpoint inhibitors is still evolving in the treatment of cancers of the gastrointestinal tract. This article examines the literature to support the use of immune checkpoint inhibitors to treat cancers of each part of the gastrointestinal system. Abstract Gastrointestinal (GI) malignancies are a heterogenous group of cancers with varying epidemiology, histology, disease course, prognosis and treatment options. Immune checkpoint inhibitors (ICIs) have changed the landscape of modern cancer treatment, though they have demonstrated survival benefit in other solid tumors more readily than in GI malignancies. This review article presents an overview of the landscape of ICI use in GI malignancies and highlights recent updates in this rapidly evolving field.
Collapse
|
76
|
Kim S, Boustani J, Vernerey D, Vendrely V, Evesque L, Francois E, Quero L, Ghiringhelli F, de la Fouchardière C, Dahan L, Bouché O, Chibaudel B, Hajbi FE, Vernet C, Rebucci-Peixoto M, Feuersinger A, Maritaz C, Borg C. Phase II INTERACT-ION study: ezabenlimab (BI 754091) and mDCF (docetaxel, cisplatin, and 5-fluorouracil) followed by chemoradiotherapy in patients with Stage III squamous cell anal carcinoma. Front Oncol 2022; 12:918499. [PMID: 36119522 PMCID: PMC9472525 DOI: 10.3389/fonc.2022.918499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background Chemoradiotherapy alone is the standard treatment for locally advanced squamous cell anal carcinoma (SCAC). However, up to 50% of patients will experience recurrence; thus, there is a need for new treatments to improve outcomes. Modified docetaxel, cisplatin and 5-fluorouracil (mDCF) is a treatment option for first-line metastatic SCAC, having shown efficacy in the Epitopes-HPV01 and -02 trials (NCT01845779 and NCT02402842). mDCF treatment also plays a role in the modulation of anti-tumor immunity, suggesting it may be a good combination partner for immunotherapy in patients with SCAC. Anti-programmed death protein-1 (PD-1) immunotherapy has been shown to be effective in metastatic SCAC. We therefore designed the INTERACT-ION study to assess the combination of mDCF with ezabenlimab (BI 754091), an anti-PD-1 antibody, followed by chemoradiotherapy, in patients with Stage III SCAC. Methods INTERACT-ION is a pivotal, open-label, single-arm phase II study in patients with treatment-naïve Stage III SCAC. Patients will receive induction treatment with mDCF (docetaxel 40 mg/m2 and cisplatin 40 mg/m2 on Day 1, 5-fluorouracil 1200 mg/m2/day for 2 days) every 2 weeks for 4 cycles and ezabenlimab (240 mg given intravenously) every 3 weeks for 3 cycles. In the absence of disease progression at 2 months, two additional cycles of mDCF and one additional cycle of ezabenlimab will be administered. Patients with radiological objective response, pathological complete/near-complete response and biological complete response will then receive an involved-node radiotherapy with intensity-modulated radiation therapy and concurrent chemotherapy, followed by ezabenlimab alone for seven cycles. All other patients will receive standard chemoradiotherapy. The primary endpoint is the clinical complete response rate 10 months after the first cycle of mDCF plus ezabenlimab. Major secondary endpoints are major pathological response and biological complete response after induction treatment. An extensive ancillary biomarker study in tumor tissue and peripheral blood will also be conducted. Discussion The addition of immunotherapy to chemotherapy is an area of active interest in metastatic anal cancer. This pivotal study will evaluate this combination in the locally advanced setting. Ancillary biomarker studies will contribute to the understanding of predictors of response or resistance to treatment. Clinical Trial Registration https://clinicaltrials.gov/ct2/show/NCT04719988, identifier NCT04719988.
Collapse
Affiliation(s)
- Stefano Kim
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, Besançon, France
- INSERM, Unit 1098, University of Bourgogne Franche-Comté, Besançon, France
- *Correspondence: Stefano Kim,
| | - Jihane Boustani
- Department of Radiotherapy, University Hospital of Besançon, Besançon, France
| | - Dewi Vernerey
- Methodology and Quality of Life in Oncology Unit, University Hospital of Besançon, Besançon, France
| | - Véronique Vendrely
- Department of Radiation Oncology, Bordeaux University Hospital, Pessac, France
| | - Ludovic Evesque
- Departement of Oncology, Centre Antoine Lacassagne, Nice, France
| | - Eric Francois
- Departement of Oncology, Centre Antoine Lacassagne, Nice, France
| | - Laurent Quero
- INSERM, Unit 1160, University of Paris, Paris, France
- Department of Radiation Oncology, Saint-Louis Hospital, APHP, Paris, France
| | | | | | - Laëtitia Dahan
- Department of Digestive Oncology, La Timone, Aix Marseille Université, Marseille, France
| | - Oliver Bouché
- Department of Digestive Oncology, Hôpital Robert Debré, Reims, France
| | - Benoist Chibaudel
- Department of Medical Oncology, Hôpital Franco-Britannique, Fondation Cognacq-Jay, Levallois-Perret, France
| | - Farid El Hajbi
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - Chloé Vernet
- Department of Digestive Oncology, Hôpital Privé Jean Mermoz, Lyon, France
| | - Magali Rebucci-Peixoto
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Alexandra Feuersinger
- Global Medical Affairs, Oncology, Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Christophe Maritaz
- Medical Affairs Department, Oncology, Boehringer Ingelheim France, Paris, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, Besançon, France
- INSERM, Unit 1098, University of Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
77
|
Rao S, Jones M, Bowman J, Tian C, Spano JP. POD1UM-303/InterAACT 2: A phase III, global, randomized, double-blind study of retifanlimab or placebo plus carboplatin–paclitaxel in patients with locally advanced or metastatic squamous cell anal carcinoma. Front Oncol 2022; 12:935383. [PMID: 36091159 PMCID: PMC9449327 DOI: 10.3389/fonc.2022.935383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022] Open
Abstract
Background Squamous carcinoma of the anal canal (SCAC) is a human papillomavirus (HPV)-driven cancer with poor prognosis in locally advanced or recurrent settings. Carboplatin–paclitaxel is the preferred first-line regimen for unresectable locally advanced or metastatic SCAC, with the reported median progression-free survival (PFS) and overall survival (OS) of 8.1 and 20.0 months, respectively. Immune checkpoint blockade (ICB) demonstrates improved survival in HPV-driven cervical and head and neck cancers. Retifanlimab (INCMGA00012) is an investigational humanized, hinge-stabilized, immunoglobulin G4κ monoclonal antibody targeting programmed cell death-1 (PD-1), with characteristics common to the ICB class. In POD1UM-202, retifanlimab showed substantial clinical activity and an expected safety profile in patients with advanced SCAC who progressed on platinum-based chemotherapy. Based on these encouraging results, POD1UM-303/InterAACT 2 (NCT04472429), a phase III, double-blind, randomized, multiregional study, investigates the addition of retifanlimab to the standard of care (SOC) carboplatin–paclitaxel in patients with inoperable locally recurrent or metastatic SCAC not previously treated with systemic chemotherapy. Methods and analysis Patients ≥18 years with inoperable locally recurrent or metastatic SCAC, measurable disease per RECIST v1.1, and no prior systemic chemotherapy or PD-(L)1-directed therapy will be enrolled and stratified by PD-L1 expression, region, and extent of disease. Patients with well-controlled human immunodeficiency virus infection are eligible. Planned enrollment is approximately 300 patients worldwide, with a 1:1 randomization to retifanlimab or placebo. Patients will receive up to six induction cycles (24 weeks) of carboplatin (area-under-the-curve 5 on day 1) and paclitaxel (80 mg/m2 on days 1, 8, and 15) every 28 days per SOC. Concurrently, retifanlimab 500 mg or placebo will be administered intravenously in a blinded fashion on day 1 of each 28-day cycle for up to 13 cycles (1 year) in the absence of unacceptable toxicity, disease progression, withdrawal of consent, loss to follow-up, or premature discontinuation. Crossover to open-label retifanlimab will be allowed for patients assigned to placebo upon verification of progression by blinded independent central radiographic review (BICR). The primary study endpoint is PFS per RECIST v1.1 by BICR. Secondary endpoints are OS, objective response rate, duration of response, disease control rate, safety, and retifanlimab pharmacokinetics. The study is currently recruiting. Clinical Trial Registration https://clinicaltrials.gov/ct2/show/NCT04472429; https://clinicaltrialsregister.eu/ctr-search/search?query=2020-000826-24
Collapse
Affiliation(s)
- Sheela Rao
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
- *Correspondence: Sheela Rao,
| | - Mark Jones
- Incyte Corporation, Wilmington, DE, United States
| | - Jill Bowman
- Incyte Corporation, Wilmington, DE, United States
| | - Chuan Tian
- Incyte Corporation, Wilmington, DE, United States
| | | |
Collapse
|
78
|
Delhiat AC, Combet-Curt V, Vendrely V. [Anal cancer: Focus on current treatment and future perspective]. Cancer Radiother 2022; 26:871-874. [PMID: 36008262 DOI: 10.1016/j.canrad.2022.06.029] [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: 06/02/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 10/15/2022]
Abstract
Anal cancer is considered a rare tumor, accounting for 6 % of digestive cancers and about 2000 new cases per year in France. It is mostly diagnosed at a localized stage. For many years, the standard of care for patients with localized disease is an association with radiotherapy and chemotherapy including Mitomycin C and 5-Fluorouracil. There weren't any major changes in the therapeutic management of these tumors despite several phase III studies. However, there is an improvement in patient prognostic. This can be explained by imaging progress, using magnetic resonance imaging and positron emission tomography-computed tomography, permitting better staging and evaluation of disease. Moreover, irradiation modalities changed because of the development of Intensity Modulated Radiotherapy. Actual research focuses on a more personalized strategy according to tumoral stages. Patients with early-stage tumors are potentially over-treated with a risk of chronic digestive toxicities. Several studies are interested in irradiation de-escalation for these patients. On the other hand, treatment results for patients with advanced tumoral stages are disappointing. It seems relevant to propose a therapeutic intensification for these patients, such as dose escalation, association with new therapies like immunotherapy or induction chemotherapy using taxans given promising results at the metastatic stage.
Collapse
Affiliation(s)
- A C Delhiat
- Service d'oncologie-radiothérapie, hôpital Haut-Lévêque, CHU de Bordeaux, Bordeaux, France
| | - V Combet-Curt
- Service d'oncologie-radiothérapie, hôpital Haut-Lévêque, CHU de Bordeaux, Bordeaux, France
| | - V Vendrely
- Service d'oncologie-radiothérapie, hôpital Haut-Lévêque, CHU de Bordeaux, Bordeaux, France; U1312-BRIC, eq BioGO, Inserm, université de Bordeaux, 33000 Bordeaux, France.
| |
Collapse
|
79
|
Mathias-Machado MC, Peixoto RD, Moniz CMV, Jácome AA. Biomarkers in Anal Cancer: Current Status in Diagnosis, Disease Progression and Therapeutic Strategies. Biomedicines 2022; 10:2029. [PMID: 36009576 PMCID: PMC9405643 DOI: 10.3390/biomedicines10082029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Squamous cell carcinoma of the anal canal (SCCA) is a rare neoplasm, but with rising incidence rates in the past few decades; it is etiologically linked with the human papillomavirus (HPV) infection and is especially prevalent in immunocompromised patients, mainly those infected with HIV. Fluoropyrimidine-based chemoradiotherapy remains the cornerstone of the treatment of non-metastatic disease, but the locally advanced disease still presents high rates of disease recurrence and systemic therapy of SCCA is an unmet clinical need. Despite sharing common molecular aspects with other HPV-related malignancies, such as cervical and head and neck cancers, SCCA presents specific epigenomic, genomic, and transcriptomic abnormalities, which suggest that genome-guided personalized therapies should be specifically designed for this disease. Actionable mutations are rare in SCCA and immune checkpoint inhibition has not yet been proven useful in an unselected population of patients. Therefore, advances in systemic therapy of SCCA will only be possible with the identification of predictive biomarkers and the subsequent development of targeted therapies or immunotherapeutic approaches that consider the unique tumor microenvironment and the intra- and inter-tumoral heterogeneity. In the present review, we address the molecular characterization of SCCA and discuss potential diagnostic, predictive and prognostic biomarkers of this complex and challenging disease.
Collapse
Affiliation(s)
- Maria Cecília Mathias-Machado
- Department of Gastrointestinal Medical Oncology, Oncoclinicas, São Paulo 04538-132, Brazil
- Department of Oncology, ICESP—Instituto do Cancer do Estado de São Paulo, University of São Paulo, São Paulo 01246-000, Brazil
| | | | - Camila Motta Venchiarutti Moniz
- Department of Oncology, ICESP—Instituto do Cancer do Estado de São Paulo, University of São Paulo, São Paulo 01246-000, Brazil
| | - Alexandre A. Jácome
- Department of Gastrointestinal Medical Oncology, Oncoclinicas, Belo Horizonte 34000-000, Brazil
| |
Collapse
|
80
|
Tarhini AA, Eads JR, Moore KN, Tatard-Leitman V, Wright J, Forde PM, Ferris RL. Neoadjuvant immunotherapy of locoregionally advanced solid tumors. J Immunother Cancer 2022; 10:jitc-2022-005036. [PMID: 35973745 PMCID: PMC9386211 DOI: 10.1136/jitc-2022-005036] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 11/25/2022] Open
Abstract
Definitive management of locoregionally advanced solid tumors presents a major challenge and often consists of a combination of surgical, radiotherapeutic and systemic therapy approaches. Upfront surgical treatment with or without adjuvant radiotherapy carries the risks of significant morbidities and potential complications that could be lasting. In addition, these patients continue to have a high risk of local or distant disease relapse despite the use of standard adjuvant therapy. Preoperative neoadjuvant systemic therapy has the potential to significantly improve clinical outcomes, particularly in this era of expanding immunotherapeutic agents that have transformed the care of patients with metastatic/unresectable malignancies. Tremendous progress has been made with neoadjuvant immunotherapy in the treatment of several locoregionally advanced resectable solid tumors leading to ongoing phase 3 trials and change in clinical practice. The promise of neoadjuvant immunotherapy has been supported by the high pathologic tumor response rates in early trials as well as the durability of these responses making cure a more achievable potential outcome compared with other forms of systemic therapy. Furthermore, neoadjuvant studies allow the assessment of radiologic and pathological responses and the access to biospecimens before and during systemic therapy. Pathological responses may guide future treatment decisions, and biospecimens allow the conduct of mechanistic and biomarker studies that may guide future drug development. On behalf of the National Cancer Institute Early Drug Development Neoadjuvant Immunotherapy Working Group, this article summarizes the current state of neoadjuvant immunotherapy of solid tumors focusing primarily on locoregionally advanced melanoma, gynecologic malignancies, gastrointestinal malignancies, non-small cell lung cancer and head and neck cancer including recent advances and our expert recommendations related to future neoadjuvant trial designs and associated clinical and translational research questions.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Jennifer R Eads
- Medicine, University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania, USA
| | - Kathleen N Moore
- Gynecologic Oncology, The University of Oklahoma Stephenson Cancer Center, Oklahoma City, Oklahoma, USA
| | | | - John Wright
- National Cancer Institute, Bethesda, Maryland, USA
| | - Patrick M Forde
- Oncology, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Robert L Ferris
- Otolaryngology and Immunology, University of Pittsburgh & UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
81
|
Welland S, deCastro T, Bathon M, Wirth TC, Reineke-Plaaß T, Saborowski M, Lehmann U, Saborowski A, Vogel A. Molecular diagnostics and therapies for gastrointestinal tumors: a real-world experience. J Cancer Res Clin Oncol 2022; 148:2137-2144. [PMID: 34436668 PMCID: PMC9293869 DOI: 10.1007/s00432-021-03774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/17/2021] [Indexed: 11/10/2022]
Abstract
PURPOSE Several targeted agents demonstrated efficacy in early clinical trials for gastrointestinal (GI) cancers, but in many cases, phase-III trials and/or approval by the European Medicines Agency (EMA) are lacking. The primary focus of this study was to assess the regulatory processes associated with use and reimbursement of off-label treatment in precision oncology and to evaluate the benefit of targeted therapy in a real-world population in Germany. METHODS Our cohort comprises 137 patients with GI cancers and is biased towards cancer entities with a high frequency of known targetable alterations, such as cholangiocarcinoma. Genetic testing was used to identify molecular targets, and therapy response was evaluated based on CT scans. RESULTS A molecular target for precision oncology was identified in 53 patients and 43 requests for cost coverage were submitted to health insurance companies. 60% of the requests received approval after initial application and another 7% after appeal. Half of the rejected requests were denied despite ESCAT IA level evidence. The median time between initiation of molecular testing and start of therapy was 75 days. 35 patients received matched targeted therapies (n = 28) or, in the case of MSI, immunotherapy (IO) (n = 7). We observed a trend in favor of molecular therapy when compared to the immediate prior treatment. CONCLUSION Relevant treatment options were identified by molecular testing in a significant subset of patients. When targeted therapies that lack EMA approval are considered, treatment initiation may be delayed by the duration of the molecular analysis and the regulatory processes.
Collapse
Affiliation(s)
- Sabrina Welland
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Tiago deCastro
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Melanie Bathon
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Christian Wirth
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Tanja Reineke-Plaaß
- Department of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Michael Saborowski
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ulrich Lehmann
- Department of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Arndt Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
82
|
Rebucci-Peixoto M, Vienot A, Adotevi O, Jacquin M, Ghiringhelli F, de la Fouchardière C, You B, Maurina T, Kalbacher E, Bazan F, Meynard G, Clairet AL, Fagnoni-Legat C, Spehner L, Bouard A, Vernerey D, Meurisse A, Kim S, Borg C, Mansi L. A Phase II Study Evaluating the Interest to Combine UCPVax, a Telomerase CD4 T H1-Inducer Cancer Vaccine, and Atezolizumab for the Treatment of HPV Positive Cancers: VolATIL Study. Front Oncol 2022; 12:957580. [PMID: 35928870 PMCID: PMC9343837 DOI: 10.3389/fonc.2022.957580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background There is a strong rational of using anti-programmed cell death protein-1 and its ligand (anti-PD-1/L1) antibodies in human papillomavirus (HPV)-induced cancers. However, anti-PD-1/L1 as monotherapy induces a limited number of objective responses. The development of novel combinations in order to improve the clinical efficacy of an anti-PD-1/L1 is therefore of interest. Combining anti-PD-1/L1 therapy with an antitumor vaccine seems promising in HPV-positive (+) cancers. UCPVax is a therapeutic cancer vaccine composed of two separate peptides derived from telomerase (hTERT, human telomerase reverse transcriptase). UCPVax is being evaluated in a multicenter phase I/II study in NSCLC (non-small cell lung cancer) and has demonstrated to be safe and immunogenic. The aim of the VolATIL study is to evaluate the combination of atezolizumab (an anti-PD-L1) and UCPVax vaccine in a multicenter phase II study in patients with HPV+ cancers. Methods Patients with HPV+ cancer (anal canal, head and neck, and cervical or vulvar), at locally advanced or metastatic stage, and refractory to at least one line of systemic chemotherapy are eligible. The primary end point is the objective response rate (ORR) at 4 months. Patients will receive atezolizumab every 3 weeks at a fixed dose of 1,200 mg in combination with the UCPVax vaccine at 1 mg subcutaneously. Discussion Anti-cancer vaccines can restore cancer-immunity via the expansion and activation of tumor-specific T cells in patients lacking pre-existing anti-tumor responses. Moreover, preclinical data showed that specific TH1 CD4 T cells sustain the quality and homing of an antigen-specific CD8+ T-cell immunity. In previous clinical studies, the induction of anti-hTERT immunity was significantly correlated to survival in patients with advanced squamous anal cell carcinoma. Thus, there is a strong rational to combine an anti-cancer hTERT vaccine and an immune checkpoint inhibitor to activate and promote antitumor T-cell immunity. This pivotal proof of concept study will evaluate the efficacy and safety of the combination of a telomerase-based TH1 inducing vaccine (UCPVax) and an anti-PD-L1 (atezolizumab) immunotherapy in HPV+ cancers, as well as confirming their synergic mechanism, and settling the basis for a new combination for future clinical trials. Clinical Trial Registration https://www.clinicaltrials.gov/, identifier NCT03946358.
Collapse
Affiliation(s)
- Magali Rebucci-Peixoto
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
- Clinical Investigational Center, CIC-1431, Centre Hospitalier Universitaire, Besançon, France
| | - Angélique Vienot
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
- Clinical Investigational Center, CIC-1431, Centre Hospitalier Universitaire, Besançon, France
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
| | - Olivier Adotevi
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
- Clinical Investigational Center, CIC-1431, Centre Hospitalier Universitaire, Besançon, France
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
| | - Marion Jacquin
- Clinical Investigational Center, CIC-1431, Centre Hospitalier Universitaire, Besançon, France
- Cancéropôle Est, Strasbourg, France
| | | | | | - Benoit You
- Department of Oncology, Hospices Civils de Lyon, Lyon, France
| | - Tristan Maurina
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
| | - Elsa Kalbacher
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
| | - Fernando Bazan
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
| | - Guillaume Meynard
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
| | - Anne-Laure Clairet
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | | | - Laurie Spehner
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
| | - Adeline Bouard
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
| | - Dewi Vernerey
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
- Methodology and Quality of Life in Oncology Unit, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Aurélia Meurisse
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
- Methodology and Quality of Life in Oncology Unit, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Stefano Kim
- Clinical Investigational Center, CIC-1431, Centre Hospitalier Universitaire, Besançon, France
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
- Department of Oncology, Sanatorio Allende, Cordoba, Argentina
| | - Christophe Borg
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
- Clinical Investigational Center, CIC-1431, Centre Hospitalier Universitaire, Besançon, France
- INSERM, EFS BFC, UMR1098 RIGHT, University of Bourgogne Franche-Comté, Besançon, France
| | - Laura Mansi
- Department of Oncology, Centre Hospitalier Universitaire, Besançon, France
| |
Collapse
|
83
|
Rao S, Anandappa G, Capdevila J, Dahan L, Evesque L, Kim S, Saunders MP, Gilbert DC, Jensen LH, Samalin E, Spindler KL, Tamberi S, Demols A, Guren MG, Arnold D, Fakih M, Kayyal T, Cornfeld M, Tian C, Catlett M, Smith M, Spano JP. A phase II study of retifanlimab (INCMGA00012) in patients with squamous carcinoma of the anal canal who have progressed following platinum-based chemotherapy (POD1UM-202). ESMO Open 2022; 7:100529. [PMID: 35816951 PMCID: PMC9463376 DOI: 10.1016/j.esmoop.2022.100529] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023] Open
Abstract
Background Locally advanced or metastatic squamous carcinoma of the anal canal (SCAC) has poor prognosis following platinum-based chemotherapy. Retifanlimab (INCMGA00012), a humanized monoclonal antibody targeting programmed death protein-1 (PD-1), demonstrated clinical activity across a range of solid tumors in clinical trials. We present results from POD1UM-202 (NCT03597295), an open-label, single-arm, multicenter, phase II study evaluating retifanlimab in patients with previously treated advanced or metastatic SCAC. Patients and methods Patients ≥18 years of age had measurable disease and had progressed following, or were ineligible for, platinum-based therapy. Retifanlimab 500 mg was administered intravenously every 4 weeks. The primary endpoint was overall response rate (ORR) by independent central review. Secondary endpoints were duration of response (DOR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety. Results Overall, 94 patients were enrolled. At a median follow-up of 7.1 months (range, 0.9-19.4 months), ORR was 13.8% [95% confidence interval (CI) 7.6% to 22.5%], with one complete response (1.1%) and 12 partial responses (12.8%). Responses were observed regardless of human immunodeficiency virus or human papillomavirus status, programmed death ligand 1 (PD-L1) expression, or liver metastases. Stable disease was observed in 33 patients (35.1%) for a DCR of 48.9% (95% CI 38.5% to 59.5%). Median DOR was 9.5 months (range, 5.6 months-not estimable). Median (95% CI) PFS and OS were 2.3 (1.9-3.6) and 10.1 (7.9-not estimable) months, respectively. Retifanlimab safety in this population was consistent with previous experience for the PD-(L)1 inhibitor class. Conclusions Retifanlimab demonstrated clinically meaningful and durable antitumor activity, and an acceptable safety profile in patients with previously treated locally advanced or metastatic SCAC who have progressed on or are intolerant to platinum-based chemotherapy. Retifanlimab (PD-1 inhibitor) monotherapy demonstrated encouraging results in patients with platinum-refractory SCAC. Clinically meaningful antitumor activity was reported with ORR of 13.8% and stable disease in 35.1%, for a DCR of 48.9%. Observed responses in advanced SCAC were durable (median 9.5 months). Acceptable safety profile consistent with that reported for the PD-(L)1 inhibitor class. Promising results warrant further investigation of retifanlimab in advanced SCAC as well as earlier stages of disease.
Collapse
Affiliation(s)
- S Rao
- The Royal Marsden, London, UK.
| | | | - J Capdevila
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Teknon-IOB, Barcelona, Spain
| | - L Dahan
- Hôpital de la Timone, Marseille, France
| | - L Evesque
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - S Kim
- Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | | | - D C Gilbert
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, UK
| | - L H Jensen
- University Hospital of Southern Denmark, Vejle, Denmark
| | - E Samalin
- Department of Digestive Oncology, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | | | - S Tamberi
- Department of Oncology/Haematology, AUSL Romagna Oncology Unit Faenza Hospital (RA), Faenza, Italy
| | - A Demols
- Department of Gastroenterology and GI Oncology, CUB Hôpital Erasme, Université Libre de Bruxelles, Anderlecht, Belgium
| | - M G Guren
- Oslo University Hospital and University of Oslo, Oslo, Norway
| | - D Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| | - M Fakih
- City of Hope Comprehensive Cancer Center, Duarte, USA
| | - T Kayyal
- Renovatio Clinical, Houston, USA
| | | | - C Tian
- Incyte Corporation, Wilmington, USA
| | | | - M Smith
- Incyte Corporation, Wilmington, USA
| | - J-P Spano
- APHP-Sorbonne University-IUC, Paris, France
| |
Collapse
|
84
|
Collienne M, Loghmani H, Heineman TC, Arnold D. GOBLET: a phase I/II study of pelareorep and atezolizumab +/- chemo in advanced or metastatic gastrointestinal cancers. Future Oncol 2022; 18:2871-2878. [PMID: 35796248 DOI: 10.2217/fon-2022-0453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Most gastrointestinal (GI) cancers have microsatellite-stable (MSS) tumors, which have an immunologically 'cold' phenotype with fewer genetic mutations, reduced immune cell infiltration and downregulated immune checkpoint proteins. These attributes make MSS tumors resistant to conventional immunotherapy including checkpoint blockade therapy. Pelareorep is a naturally occurring, nongenetically modified reovirus. Upon intravenous administration, pelareorep selectively kills tumor cells and promotes several immunologic changes that prime tumors to respond to checkpoint blockade therapy. Given its demonstrated synergy with checkpoint blockade, as well as its encouraging efficacy in prior GI cancer studies, pelareorep plus atezolizumab will be evaluated in the GOBLET study in multiple GI cancer indications.
Collapse
Affiliation(s)
- Maike Collienne
- Asklepios Tumorzentrum Hamburg, AK Altona, Oncology, Hematology, Palliative Care, Rheumatology, Paul-Ehrlich-Straße 1, Hamburg, 22763, Germany
| | - Houra Loghmani
- Oncolytics Biotech Inc., Suite 804, 322 - 11th Avenue SW, Calgary, Alberta, T2R 0C5, Canada
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Oncology, Hematology, Palliative Care, Rheumatology, Paul-Ehrlich-Straße 1, Hamburg, 22763, Germany
| |
Collapse
|
85
|
Gameiro SR, Strauss J, Gulley JL, Schlom J. Preclinical and clinical studies of bintrafusp alfa, a novel bifunctional anti-PD-L1/TGFβRII agent: Current status. Exp Biol Med (Maywood) 2022; 247:1124-1134. [PMID: 35473390 PMCID: PMC9335510 DOI: 10.1177/15353702221089910] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Bintrafusp alfa (anti-PD-L1/TGFβRII) is a first-in-class bifunctional agent designed to act both as a checkpoint inhibitor and as a "trap" for TGFβ in the tumor microenvironment (TME). This article is designed to review the preclinical studies interrogating the mode of action of bintrafusp alfa and to present a comprehensive overview of recent bintrafusp alfa clinical studies. Preclinical studies have demonstrated that bintrafusp alfa immune-mediating and antitumor activity can be enhanced by combining it with a human papillomavirus (HPV) therapeutic cancer vaccine, a tumor-targeting interleukin 12 (IL-12) immunocytokine and/or an IL-15 superagonist. The importance of TGFβ in HPV-associated malignancies is also reviewed. The clinical studies reviewed span extended phase I cohorts in patients with a spectrum of malignancies, two randomized phase II studies in lung and one in biliary tract cancers in which bintrafusp alfa did not demonstrate superiority over standard-of-care therapies, and provocative results in patients with HPV-associated malignancies, where as a monotherapy, bintrafusp alfa has shown response rates of 35%, compared to overall response rate (ORR) of 12-24% seen with other Food and Drug Administration (FDA)-approved or standard-of-care agents. This article also reviews preliminary phase II study results of patients with HPV+ malignancies employing bintrafusp alfa in combination with an HPV therapeutic vaccine and a tumor-targeting IL-12 immunocytokine in which the combination therapy outperforms standard-of-care therapies in both checkpoint naïve and checkpoint refractory patients. This review thus provides an example of the importance of conducting clinical studies in an appropriate patient population - in this case, exemplified by the role of TGFβ in HPV-associated malignancies. This review also provides preclinical and preliminary clinical study results of the combined use of multiple immune-modulating agents, each designed to engage different immune components and tumor cells in the TME.
Collapse
Affiliation(s)
- Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julius Strauss
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
86
|
Editorial: 'All you need is immunotherapy': a new treatment paradigm in sight for early-stage, mismatch repair-deficient/microsatellite instability high gastrointestinal cancers? Curr Opin Oncol 2022; 34:379-381. [PMID: 35837708 DOI: 10.1097/cco.0000000000000856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
87
|
Lin YT, Liu TX, Chen J, Wang C, Chen Y. Cost-Effectiveness of Nivolumab Immunotherapy vs. Paclitaxel or Docetaxel Chemotherapy as Second-Line Therapy in Advanced Esophageal Squamous Cell Carcinoma in China. Front Public Health 2022; 10:923619. [PMID: 35844891 PMCID: PMC9277084 DOI: 10.3389/fpubh.2022.923619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/09/2022] [Indexed: 12/24/2022] Open
Abstract
This study aimed to evaluate and compare nivolumab's cost-effectiveness with chemotherapy in patients with advanced esophageal squamous cell carcinoma from the Chinese healthcare system perspective. To this end, the researchers utilized a partitioned survival model with three mutually exclusive health stages. The characteristics of the patients used as inclusion and exclusion criteria in this model were the same as those used for patients with advanced esophageal squamous cell carcinoma in the ATTRACTION-3 study. The ATTRACTION-3 trial, which took place between January 7, 2016 and November 12, 2018, also yielded important clinical data. Data on medical and economic preferences were collected from real-world clinical practices. Costs, quality-adjusted life years, and incremental cost-effectiveness ratio were calculated for the two therapy options. The model uncertainty was investigated using a deterministic and probabilistic sensitivity analysis. When compared to chemotherapy, nivolumab was linked with an increase of 0.28 quality-adjusted life years with an increased cost of US$ 36,956.81 per patient in the base case analysis of a hypothetical sample of 419 patients. The incremental cost-effectiveness ratio in the deterministic sensitivity analysis was US$ 132,029.46/quality-adjusted life year, with a 48.02% probability of being cost-effective at willingness-to-pay thresholds of US$ 132,029.22/quality-adjusted life year. The incremental cost-effectiveness ratio remained greater than US$ 80,000/quality-adjusted life year in the deterministic sensitivity analyses. To be more cost-effective and remain below the threshold of 37,653 US$/quality-adjusted life year, which the Chinese population can afford, nivolumab's price would have to be lowered sharply by 53.50%. Nivolumab is clinically beneficial but not cost-effective when compared to chemotherapy. A substantial reduction in nivolumab's drug acquisition cost would be necessary to make it cost-effective for immunotherapy.
Collapse
Affiliation(s)
- Ying-tao Lin
- Department of Drug Clinical Trial Administration Office, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Tian-xiu Liu
- Department of Thoracic Radiotherapy, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jian Chen
- Department of Surgical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Chang Wang
- Department of Medical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Ying Chen
- Department of Drug Clinical Trial Administration Office, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
88
|
Principe DR, Cataneo JL, Timbers KE, Koch RM, Valyi-Nagy K, Mellgren A, Rana A, Gantt G. Leukocyte subtyping predicts for treatment failure and poor survival in anal squamous cell carcinoma. BMC Cancer 2022; 22:697. [PMID: 35751111 PMCID: PMC9229146 DOI: 10.1186/s12885-022-09742-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Anal squamous cell carcinoma (SCC) generally carries a favorable prognosis, as most tumors are highly sensitive to standard of care chemoradiation. However, outcomes are poor for the 20–30% of patients who are refractory to this approach, and many will require additional invasive procedures with no guarantee of disease resolution. Methods To identify the patients who are unlikely to respond to the current standard of care chemoradiation protocol, we explored a variety of objective clinical findings as a potential predictor of treatment failure and/or mortality in a single center retrospective study of 42 patients with anal SCC. Results Patients with an increase in total peripheral white blood cells (WBC) and/or neutrophils (ANC) had comparatively poor clinical outcomes, with increased rates of death and treatment failure, respectively. Using pre-treatment biopsies from 27 patients, tumors with an inflamed, neutrophil dominant stroma also had poor therapeutic responses, as well as reduced overall and disease-specific survival. Following chemoradiation, we observed uniform reductions in nearly all peripheral blood leukocyte subtypes, and no association between peripheral white blood cells and/or neutrophils and clinical outcomes. Additionally, post-treatment biopsies were available from 13 patients. In post-treatment specimens, patients with an inflamed tumor stroma now demonstrated improved overall and disease-specific survival, particularly those with robust T-cell infiltration. Conclusions Combined, these results suggest that routinely performed leukocyte subtyping may have utility in risk stratifying patients for treatment failure in anal SCC. Specifically, pre-treatment patients with a high WBC, ANC, and/or a neutrophil-dense tumor stroma may be less likely to achieve complete response using the standard of care chemoradiation regimen, and may benefit from the addition of a subsequent line of therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09742-7.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA.,Department of Surgery, University of Illinois at Chicago, IL, Chicago, USA
| | - Jose L Cataneo
- Department of Surgery, University of Illinois at Chicago, IL, Chicago, USA
| | - Kaytlin E Timbers
- Department of Surgery, University of Illinois at Chicago, IL, Chicago, USA
| | - Regina M Koch
- Department of Surgery, University of Illinois at Chicago, IL, Chicago, USA
| | - Klara Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago, IL, Chicago, USA
| | - Anders Mellgren
- Department of Surgery, Division of Colorectal Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, IL, Chicago, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Gerald Gantt
- Department of Surgery, Division of Colorectal Surgery, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
89
|
Rogers JE, Leung M, Johnson B. Metastatic or Locally Recurrent Anal Squamous Cell Carcinoma (SCAC): Current Clinical Trial Landscape and Novel Approaches. Cancer Manag Res 2022; 14:2065-2077. [PMID: 35761823 PMCID: PMC9233494 DOI: 10.2147/cmar.s331429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022] Open
Abstract
Anal squamous cell carcinoma (SCAC) is a human papillomavirus (HPV) driven malignancy. Given inadequate HPV-vaccination rates, SCAC will continue to be a public health concern. SCAC is commonly diagnosed in the local or locoregional setting in which definitive chemoradiation provides the opportunity for cure and has high control rates. A minority of patients will develop recurrence or present with metastatic SCAC. Given the rarity of this disease, research has lagged compared to many other solid tumors. Historically, treatment has been based on extrapolating management approaches from more common squamous cell carcinoma malignancies and/or small case series or case reports. Fortunately, dedicated prospective clinical trial investigation in the advanced setting has emerged in recent years. Here, we review the current strategies for treatment along with remaining challenges and viable next steps for the management of metastatic SCAC.
Collapse
Affiliation(s)
- Jane E Rogers
- U.T. M.D. Anderson Cancer Center, Pharmacy Clinical Programs, Houston, TX, USA
| | - Michael Leung
- U.T. M.D. Anderson Cancer Center, Pharmacy Clinical Programs, Houston, TX, USA
| | - Benny Johnson
- U.T. M.D. Anderson Cancer Center, Department of Gastrointestinal Medical Oncology, Houston, TX, USA
| |
Collapse
|
90
|
Guven DC, Stephen B, Sahin TK, Cakir IY, Erul E, Aksoy S. The efficacy of immune checkpoint inhibitors in rare tumors: A systematic review of published clinical trials. Crit Rev Oncol Hematol 2022; 174:103700. [PMID: 35533815 DOI: 10.1016/j.critrevonc.2022.103700] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/22/2022] [Accepted: 05/02/2022] [Indexed: 02/08/2023] Open
Abstract
The immune checkpoint inhibitors (ICIs) entered treatment algorithms in most tumors. However, the data on the efficacy is limited in rare tumors with no phase III studies. We systemically reviewed the clinical trials evaluating the ICI efficacy in rare tumors and included a total of 47 clinical trials in this review. The ICIs demonstrated over 30% response rates in Merkel cell carcinoma and squamous cell carcinoma of the skin and became the standard of care. Additionally, the ICI efficacy was promising in thymic epithelial tumors and gestational trophoblastic neoplasia. In contrast, the ICI efficacy is limited in most sarcomas, germ cell tumors and low-grade neuroendocrine tumors. The ICI efficacy seemed to be improved with combinations targeting tumor microenvironment in sarcomas. The available evidence on ICI efficacy in rare tumors denote a need for better patient selection and novel combination strategies to improve outcomes.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey.
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Taha Koray Sahin
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ibrahim Yahya Cakir
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Enes Erul
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
91
|
Eng C, Ciombor KK, Cho M, Dorth JA, Rajdev LN, Horowitz DP, Gollub MJ, Jácome AA, Lockney NA, Muldoon RL, Washington MK, O'Brian BA, Benny A, Lebeck Lee CM, Benson AB, Goodman KA, Morris VK. Anal Cancer: Emerging Standards in a Rare Rare Disease. J Clin Oncol 2022; 40:2774-2788. [PMID: 35649196 DOI: 10.1200/jco.21.02566] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The social stigma surrounding an anal cancer diagnosis has traditionally prevented open discussions about this disease. However, as recent treatment options and an increasing rate of diagnoses are made worldwide, awareness is growing. In the United States alone, 9,090 individuals were expected to be diagnosed with anal cancer in 2021. The US annual incidence of squamous cell carcinoma of the anus continues to increase by 2.7% yearly, whereas the mortality rate increases by 3.1%. The main risk factor for anal cancer is a human papillomavirus infection; those with chronic immunosuppression are also at risk. Patients with HIV are 19 times more likely to develop anal cancer compared with the general population. In this review, we have provided an overview of the carcinoma of the anal canal, the role of screening, advancements in radiation therapy, and current trials investigating acute and chronic treatment-related toxicities. This article is a comprehensive approach to presenting the existing data in an effort to encourage continuous international interest in anal cancer.
Collapse
Affiliation(s)
- Cathy Eng
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Kristen K Ciombor
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - May Cho
- Division of Hematology and Oncology, Department of Medicine, University of California- Irvine School of Medicine, Irvine, CA
| | - Jennifer A Dorth
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Lakshmi N Rajdev
- Division for Hematology and Oncology, Department of Medicine, Northwell Health/Lenox Hill Hospital, New York, NY
| | - David P Horowitz
- Department of Radiation Oncology, New York Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY
| | - Marc J Gollub
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Alexandre A Jácome
- OncoBio Comprehensive Cancer Center, Department of Gastrointestinal Medical Oncology, Nova Lima, Brazil
| | - Natalie A Lockney
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Roberta L Muldoon
- Division of Colon and Rectal Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Mary Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Brittany A O'Brian
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Amala Benny
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Cody M Lebeck Lee
- VA Tennessee Valley Healthcare System, Department of Internal Medicine, Nashville, TN
| | - Al B Benson
- Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Van Karlyle Morris
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
92
|
Li Y, Zang Y, Fan T, Li Z, Li A, Lv W, Wang Q, Li Q, Li Y, Li Q, Sun Z, Teng H. Transcriptomic signatures associated with autoimmune thyroiditis in papillary thyroid carcinoma and cancer immunotherapy-induced thyroid dysfunction. Comput Struct Biotechnol J 2022; 20:2391-2401. [PMID: 35664236 PMCID: PMC9125670 DOI: 10.1016/j.csbj.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/22/2022] [Accepted: 05/12/2022] [Indexed: 12/03/2022] Open
Abstract
Up to 20% of patients treated with anti-PD-1/PD-L1 inhibitors suffered from thyroid dysfunctions, yet the mediators associated with their occurrence remain unclear. The increasing coincidence of papillary thyroid carcinoma (PTC) with Hashimoto thyroiditis (HT) and the high vulnerability of thyroid to immunotherapy motivated us to discover the similarities and their underlying transcriptomic basis. Clinical characteristics analysis of 468 PTC patients from two independent cohorts and meta-analysis of 22,155 PTC patients unveiled a strong negative association between HT and recurrence in PTC patients. Transcriptome analysis of both cohorts showed PTC patients with HT were enriched in macrophages, CD8+ and CD4+ cytotoxic T cells, which was further validated by single-cell transcriptome analysis of 17,438 cells from PTC patients, and CD8+ T cells were correlated with disease-free survival of PTC patients. In both cohorts and single-cell dataset, elevated expression of PD-1-related genes was observed in the HT group, and CD3D appeared to be a target for enhancing the activation of CD8+ T cells. Correlation analysis of 3,318 thyroid adverse events from 39,123 patients across 24 tumor types and molecular signatures demonstrated similar signatures associated with autoimmune thyroiditis in PTC and thyroid immune-related adverse events (irAEs), and several multi-omics signatures, including signatures of CD8A and CD8+ T cells, showed positive associations with the odds ratio of thyroid irAEs. Our results unveil shared molecular signatures underlying thyroid dysfunction between patients receiving immunotherapies and PTC patients suffering from HT, which may shed light on managing the adverse events during cancer immunotherapy.
Collapse
Affiliation(s)
- Yi Li
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yue Zang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Tianda Fan
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhaochen Li
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Anzi Li
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Wei Lv
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Qingqing Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Qinglan Li
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Yuanyuan Li
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Quan Li
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Zhongsheng Sun
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- IBMC-BGI Center, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Huajing Teng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
93
|
Marabelle A, Cassier PA, Fakih M, Kao S, Nielsen D, Italiano A, Guren TK, van Dongen MGJ, Spencer K, Bariani GM, Ascierto PA, Santoro A, Shah M, Asselah J, Iqbal S, Takahashi S, Piha-Paul SA, Ott PA, Chatterjee A, Jin F, Norwood K, Delord JP. Pembrolizumab for previously treated advanced anal squamous cell carcinoma: results from the non-randomised, multicohort, multicentre, phase 2 KEYNOTE-158 study. Lancet Gastroenterol Hepatol 2022; 7:446-454. [PMID: 35114169 PMCID: PMC12012850 DOI: 10.1016/s2468-1253(21)00382-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Outcomes in advanced anal squamous cell carcinoma are poor, with few treatment options and controlled clinical trials. We evaluated the efficacy and safety of pembrolizumab in patients with advanced anal squamous cell carcinoma (cohort A) from the phase 2 KEYNOTE-158 study. METHODS Eligible patients enrolled in the ongoing non-randomised, multicohort, multicentre, phase 2 KEYNOTE-158 study, which was done across 38 centres worldwide, were aged 18 years or older; had histologically or cytologically confirmed advanced or metastatic anal squamous cell carcinoma; had previous failure of or intolerance to standard therapy or no standard therapy options; and had a PD-L1-evaluable tissue sample. Patients received pembrolizumab 200 mg intravenously every 3 weeks for 2 years, or until disease progression, unacceptable toxicity, investigator's decision to withdraw the patient from the study, or withdrawal of patient consent. The primary endpoint was objective response, as assessed by Response Evaluation Criteria in Solid Tumors version 1.1. Efficacy and safety analyses included all patients who received at least one dose of pembrolizumab. The trial is registered with ClinicalTrials.gov, NCT02628067. FINDINGS Between March 3, 2016, and July 23, 2018, 163 patients were screened, of whom 112 were enrolled and treated in the anal cancer cohort. 91 (81%) patients were female, 104 (93%) had M1 disease, and 75 (67%) had PD-L1-positive tumours. The median time from first dose to data cutoff (June 27, 2019) was 34·7 months (IQR 32·5-36·4). 12 (11%, 95% CI 6-18) patients had an objective response, including 11 (15%, 8-25) of 75 patients with PD-L1-positive tumours and one (3%; 0-17) of 30 patients with PD-L1-negative tumours. 68 (61%) patients had treatment-related adverse events (20 [18%] patients had grade 3-4 adverse events), the most common of which were fatigue (17 patients), diarrhoea (13), hypothyroidism (13), and nausea (13). Serious treatment-related adverse events occurred in 12 (11%) patients. 25 (22%) patients had immune-mediated adverse events, and one (1%) had an infusion reaction. There were no treatment-related deaths. INTERPRETATION Pembrolizumab monotherapy is a possible treatment option with a favourable benefit-risk ratio for patients with previously treated advanced anal squamous cell carcinoma who have no alternative satisfactory treatment options. FUNDING Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Aurelien Marabelle
- Gustave Roussy, INSERM U1015, Université Paris Saclay, Villejuif, France.
| | | | - Marwan Fakih
- City of Hope National Medical Center, Duarte, CA, USA
| | - Steven Kao
- Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Dorte Nielsen
- Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Antoine Italiano
- Early Phase Trials Unit, Institut Bergonié and University of Bordeaux, Bordeaux, France
| | | | | | - Kristen Spencer
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Armando Santoro
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Italy; IRCCS Humanitas Research Hospital-Humanitas Cancer Center, Rozzano, Italy
| | - Manisha Shah
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Syma Iqbal
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Shunji Takahashi
- Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | | | - Fan Jin
- Merck & Co, Kenilworth, NJ, USA
| | | | | |
Collapse
|
94
|
Lièvre A. Anti-PD-1 for pretreated advanced anal carcinoma: which patients will benefit? Lancet Gastroenterol Hepatol 2022; 7:384-386. [PMID: 35114168 DOI: 10.1016/s2468-1253(22)00002-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Affiliation(s)
- Astrid Lièvre
- Department of Gastroenterology, Pontchaillou University Hospital, Rennes University, INSERM U1242, Rennes, France.
| |
Collapse
|
95
|
Tsai YT, Strauss J, Toney NJ, Jochems C, Venzon DJ, Gulley JL, Schlom J, Donahue RN. Immune correlates of clinical parameters in patients with HPV-associated malignancies treated with bintrafusp alfa. J Immunother Cancer 2022; 10:jitc-2022-004601. [PMID: 35418484 PMCID: PMC9014099 DOI: 10.1136/jitc-2022-004601] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2022] [Indexed: 12/20/2022] Open
Abstract
Purpose Bintrafusp alfa is a bifunctional agent consisting of an anti-human PD-L1 antibody linked to two TGFβRII. It is designed to act both as a checkpoint inhibitor and to ‘trap’ TGFβ in the tumor microenvironment. Phase I and II clinical studies demonstrated clinical activity in patients with a range of human papillomavirus (HPV)-associated cancers. The purpose of the studies reported here was the interrogation of various aspects of the peripheral immunome in patients with HPV-associated cancers, both prior to and early in the treatment regimen of bintrafusp alfa to better understand the mode of action of the agent and to help define which patients are more likely to benefit from bintrafusp alfa treatment. Patients and methods The peripheral immunome of patients (n=65) with HPV+ malignancies was analyzed both prior to treatment with bintrafusp alfa and day 14 post-treatment for levels and changes in (1) 158 different immune cell subsets, (2) multiple plasma soluble factors including analytes reflecting immune stimulatory and inhibitory status, (3) complete blood counts, and in a subset of patients (4) TCR diversity and (5) HPV-specific T-cell responses. Results Interrogation of the peripheral immunome prior to bintrafusp alfa treatment revealed several factors that associated with clinical response, including (1) higher levels of sCD27:sCD40L ratios, (2) lower levels of TGFβ1 and 12 additional factors associated with tumor mesenchymalization, and (3) higher CD8+ T cell:MDSC ratios. Analysis at 2 weeks post bintrafusp alfa revealed that eventual clinical responders had fewer increases in IL-8 levels and the neutrophil to lymphocyte ratio, and higher levels of HPV-16 specific CD8+ T cells. This study also provided information concerning differences in the peripheral immunome for patients who were naïve versus refractory to prior checkpoint inhibition therapy. While preliminary, two multivariate models developed predicted clinical benefit with 76%–91% accuracy. Conclusions These studies add insight into the mechanism of action of bintrafusp alfa and provide evidence that the interrogation of both cellular and soluble components of the peripheral immunome of patients with HPV-associated malignancies, either prior to or early in the therapeutic regimen, can provide information as to which patients are more likely to benefit with bintrafusp alfa therapy.
Collapse
Affiliation(s)
- Yo-Ting Tsai
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Julius Strauss
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Nicole J Toney
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - David J Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
96
|
Immunotherapy in advanced anal cancer: Is the beginning of a new era? Cancer Treat Rev 2022; 105:102373. [DOI: 10.1016/j.ctrv.2022.102373] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022]
|
97
|
Martinez-Cannon BA, Perez ACT, Hincapie-Echeverri J, Roy M, Marinho J, Buerba GA, Akagunduz B, Li D, Soto-Perez-de-Celis E. Anal cancer in older adults: A Young International Society of Geriatric Oncology review paper. J Geriatr Oncol 2022; 13:914-923. [DOI: 10.1016/j.jgo.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
|
98
|
Yan FF, Jiang Q, Ru B, Fei XJ, Ruan J, Zhang XC. Metastatic urothelial carcinoma harboring ERBB2/3 mutations dramatically respond to chemotherapy plus anti-PD-1 antibody: A case report. World J Clin Cases 2022; 10:2497-2503. [PMID: 35434068 PMCID: PMC8968593 DOI: 10.12998/wjcc.v10.i8.2497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/25/2021] [Accepted: 01/29/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) targeting the programmed death (PD)-1 pathway have substantially changed the clinical management of metastatic urothelial carcinoma (mUC); however, the response rate remains low. There are ongoing efforts to identify robust biomarkers that can effectively predict the treatment response to ICIs. Previous studies have suggested that ERBB2/3 mutations are associated with the efficacy of ICIs in gallbladder carcinoma.
CASE SUMMARY We present a 59-year-old man with mUC harboring ERBB2/3 mutations (in-frame insertion of ERBB2 and ERBB3 amplification), negative PD-ligand 1 expression, and low tumor mutation burden. He received anti-PD-1 antibodies and paclitaxel as second-line treatment. After two cycles of treatment, the lung metastases had significantly shrunk, achieving good partial remission. After six cycles of combination therapy, the patient received sindilimab 200 mg once every 3 wk as maintenance monotherapy. At the last follow-up, the patient continued to exhibit a partial response and progression-free survival for as long as 19 mo.
CONCLUSION ERBB2/3 mutations may represent a predictive biomarker for selecting a subgroup of mUC patients who will benefit from ICIs.
Collapse
Affiliation(s)
- Fei-Fei Yan
- Department of Medical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Qi Jiang
- Department of Medical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Bin Ru
- Department of Pain Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Xiao-Jie Fei
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine Hangzhou 313000, Zhejiang Province, China
| | - Jian Ruan
- Department of Medical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Xiao-Chen Zhang
- Department of Medical Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
99
|
Tchelebi LT, Eng C, Messick CA, Hong TS, Ludmir EB, Kachnic LA, Zaorsky NG. Current treatment and future directions in the management of anal cancer. CA Cancer J Clin 2022; 72:183-195. [PMID: 34847242 DOI: 10.3322/caac.21712] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/15/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
Although rare, the rate of squamous cell carcinoma of the anus (SCCA) is rising globally. Most patients present with nonmetastatic disease and are curable with appropriate treatment, which has evolved significantly over the last several decades. Before the 1970s, SCCA was managed with radical surgery, resulting in a permanent colostomy. Researchers found that preoperative treatment with chemotherapy and concurrent radiation could achieve a pathologic complete response. After this observation, definitive therapy shifted from radical surgery to sphincter-preserving chemoradiation. Investigations into the necessity of chemotherapy and the optimal regimen found that chemotherapy with mitomycin-C and 5-fluorouracil is required for cure. Further studies evaluating the addition of induction or maintenance chemotherapy, monoclonal antibody therapy, or higher radiation doses have demonstrated no significant benefit to disease control. Advanced radiation delivery with intensity-modulated radiotherapy techniques is now considered the standard of care because of its prospectively determined, favorable acute toxicity profile compared with 3-dimensional conformal radiation. It is important to note that chemoradiation treatment response may be slow (up to 26 weeks) and should be assessed through serial clinical examinations. Today, surgical management of SCCA is reserved only for the lowest risk, early stage tumors or for recurrent/persistent disease. Current studies are evaluating radiation dose de-escalation in early stage disease and radiation dose escalation and the addition of immune checkpoint inhibitors in locally advanced cancers. In reviewing how and why modern-day treatment of SCCA was established, the objective of this report is to reenforce adherence to current treatment paradigms to assure the best possible outcomes for patients.
Collapse
Affiliation(s)
- Leila T Tchelebi
- Department of Radiation Medicine, Zucker School of Medicine, Hempstead, New York
- Department of Radiation Medicine, Northwell Health Cancer Institute, Mount Kisco, New York
| | - Cathy Eng
- Department of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Craig A Messick
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ethan B Ludmir
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lisa A Kachnic
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve School of Medicine, Cleveland, Ohio
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, Pennsylvania
| |
Collapse
|
100
|
Xiao W, Yuan Y, Wang S, Liao Z, Cai P, Chen B, Zhang R, Wang F, Zeng Z, Gao Y. Neoadjuvant PD-1 Blockade Combined With Chemotherapy Followed by Concurrent Immunoradiotherapy in Locally Advanced Anal Canal Squamous Cell Carcinoma Patients: Antitumor Efficacy, Safety and Biomarker Analysis. Front Immunol 2022; 12:798451. [PMID: 35095878 PMCID: PMC8794813 DOI: 10.3389/fimmu.2021.798451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
Background Anal canal squamous cell carcinoma (ACSCC) is an exceedingly rare malignant neoplasm with challenges in sphincter preservation, treatment toxicities and long-term survival. Little is known concerning the activity of PD-1 antibodies in locally advanced ACSCC. This study reports on the efficacy and toxicities of a neoadjuvant PD-1 blockade combined with chemotherapy followed by concurrent immunoradiotherapy in ACSCC patients, and describes biomarkers expression and mutation signatures. Methods In this cohort study, patients were treated as planned, including four cycles of neoadjuvant PD-1 antibody toripalimab combined with docetaxol and cisplatin, followed by radiotherapy and two cycles of concurrent toripalimab. Multiplex immunofluorescence staining (mIHC) with PD-L1, CD8, CD163, Pan-Keratin and DAPI was performed with the pretreatment tumor tissue. Whole exome sequencing was performed for the primary tumor and peripheral blood mononuclear cells. The primary endpoint was the complete clinical response (cCR) rate at 3 months after overall treatment. Acute and late toxicities graded were assessed prospectively. Results Five female patients with a median age of 50 years old (range, 43-65 years old), finished treatment as planned. One patient had grade 3 immune related dermatitis. Two patients had grade 3 myelosuppression during neoadjuvant treatment. No severe radiation-related toxicities were noted. Four patients with PD-L1 expression >1% achieved a cCR after neoadjuvant treatment. and the other patient with negative PD-L1 expression also achieved a cCR at 3 months after radiotherapy. All the patients were alive and free from disease and had a normal quality of life, with 19.6-24 months follow up. Inconsistent expression of PD-L1 and CD163 was detected in 3 and 5 patients, respectively. TTN, POLE, MGAM2 were the top mutation frequencies, and 80 significant driver genes were identified. Pathway analysis showed enrichment of apoptosis, Rap1, Ras, and pathways in cancer signaling pathways. Eight significantly deleted regions were identified. Conclusions This small cohort of locally advanced ACSCC patients had quite satisfactory cCR and sphincter preservation rate, after neoadjuvant PD-1 antibody toripalimab combined with chemotherapy followed by concurrent immunoradiotherapy, with mild acute and long-term toxicities.
Collapse
Affiliation(s)
- WeiWei Xiao
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yan Yuan
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - SuiHai Wang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhidong Liao
- Department of Pathology, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| | - PeiQiang Cai
- Departments of Medical Imaging and Interventional Radiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - BaoQing Chen
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Rong Zhang
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Fang Wang
- Department of Molecular Diagnosis, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - ZhiFan Zeng
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - YuanHong Gao
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|