101
|
Jin C, Zhu X, Huang X, Gong T, Wei Z, You J. Efficacy and safety of PD-1/PD-L1 and CTLA-4 immune checkpoint inhibitors in colorectal cancer: a meta-analysis. J Comp Eff Res 2022; 11:203-212. [PMID: 35023361 DOI: 10.2217/cer-2021-0134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aims: To evaluate the efficacy and safety of PD-1/PD-L1 and/or CTLA-4 inhibitors in the treatment of colorectal cancer (CRC) by meta-analysis. Methods: Electronic databases were searched. Eligible studies included investigations of efficacy and safety of anti-PD-1/PD-L1 or anti-CTLA-4 agents in patients with CRC. Corresponding indicators were calculated. Results: A total of 15 articles were included. The pooled objective response rate, overall survival rate, progression-free survival rate and adverse event rate were 33, 56, 46 and 59%, respectively. The objective response rates for CRC with deficient mismatch repair and CRC with proficient mismatch repair were 43 and 3%, respectively, in patients treated with PD-1 inhibitors. Conclusion: The authors' study indicates that PD-1/PD-L1 inhibitors manifest promising clinical responses in the treatment of CRC with deficient mismatch repair with acceptable treatment-related adverse events.
Collapse
Affiliation(s)
- Chunhui Jin
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Xiaodan Zhu
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Xiaona Huang
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Tingjie Gong
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Zhipeng Wei
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Jianliang You
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| |
Collapse
|
102
|
Abu Khalaf S, Dandachi D, Granwehr BP, Rodriguez-Barradas MC. Cancer immunotherapy in adult patients with HIV. J Investig Med 2022; 70:883-891. [PMID: 35086858 DOI: 10.1136/jim-2021-002205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 11/03/2022]
Abstract
The availability of antiretroviral therapy (ART) has increased the life expectancy of people with HIV (PWH) and reduced the incidence of AIDS-associated malignancies, yet PWH have a significantly increased incidence of malignancy and less favorable outcomes of cancer treatment compared with the general population.Immunotherapy has revolutionized cancer therapy, becoming the standard of care for various malignancy treatments. However, PWH are an underserved population with limited access to clinical trials and cancer treatment.This review of the available evidence on different classes of cancer immunotherapy in PWH is mostly based on case reports, case series, but few prospective studies and clinical trials due to the exclusion of PWH from most oncologic clinical trials. The results of the available evidence support the safety of immunotherapy in PWH. Immunotherapy has similar effectiveness in PWH, an acceptable toxicity profile, and has no clinically significant impact on HIV viral load and CD4-T cell count. In addition, there is no reported change in the incidence of opportunistic infections and other complications for PWH with well-controlled viremia.This review aims to briefly summarize the current state of immunotherapy in cancer, guide clinicians in the management of immunotherapy in cancer PWH, and encourage the inclusion of PWH in clinical trials of cancer immunotherapy.
Collapse
Affiliation(s)
- Suha Abu Khalaf
- Department of Medicine, Division of Infectious Diseases, University of Missouri System, Columbia, Missouri, USA
| | - Dima Dandachi
- Department of Medicine, Division of Infectious Diseases, University of Missouri System, Columbia, Missouri, USA
| | - Bruno P Granwehr
- Department of Medicine, Division of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maria C Rodriguez-Barradas
- Infectious Diseases Section, Michael E DeBakey VAMC, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
103
|
Evaluation of prognostic factors after primary chemoradiotherapy of anal cancer: A multicenter study of the German Cancer Consortium-Radiation Oncology Group (DKTK-ROG). Radiother Oncol 2022; 167:233-238. [PMID: 34999135 DOI: 10.1016/j.radonc.2021.12.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/21/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Prognosis after chemoradiotherapy (CRT) for anal squamous cell carcinoma (ASCC) shows marked differences among patients according to TNM subgroups, however individualized risk assessment tools to better stratify patients for treatment (de-) escalation or intensified follow-up are lacking in ASCC. MATERIALS AND METHODS Patients' data from eight sites of the German Cancer Consortium - Radiation Oncology Group (DKTK-ROG), comprising a total of 605 patients with ASCC, treated with standard definitive CRT with 5-FU/Mitomycin C or Capecitabine/Mitomycin C between 2004-2018, were used to evaluate prognostic factors based on Cox regression models for disease-free survival (DFS). Evaluated variables included age, gender, Karnofsky performance score (KPS), HIV-status, T-category, lymph node status and laboratory parameters. Multivariate cox models were separately constructed for the whole cohort and the subset of patients with early-stage (cT1-2 N0M0) tumors. RESULTS After a median follow-up of 46 months, 3-year DFS for patients with early-stage ASCC was 84.9%, and 67.1% for patients with locally-advanced disease (HR 2.4, p < 0.001). T-category (HR vs. T1: T2 2.02; T3 2.11; T4 3.03), N-category (HR versus N0: 1.8 for N1-3), age (HR 1.02 per year), and KPS (HR 0.8 per step) were significant predictors for DFS in multivariate analysis in the entire cohort. The model performed with a C-index of 0.68. In cT1-2N0 patients, T-category (HR 2.14), HIV status (HR 2.57), age (1.026 per year), KPS (HR 0.7 per step) and elevated platelets (HR 1.3 per 100/nl) were associated with worse DFS (C-index of 0.7). CONCLUSION Classical clinicopathologic parameters like T-category, N-category, age and KPS remain to be significant prognostic factors for DFS in patients treated with contemporary CRT for ASCC. HIV and platelets were significantly associated with worse DFS in patients with early stage ASCC.
Collapse
|
104
|
Second-line treatment after docetaxel, cisplatin and 5-fluorouracil in metastatic squamous cell carcinomas of the anus. Pooled analysis of prospective Epitopes-HPV01 and Epitopes-HPV02 studies. Eur J Cancer 2022; 162:138-147. [PMID: 34995900 DOI: 10.1016/j.ejca.2021.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/01/2021] [Accepted: 11/14/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Squamous cell carcinoma of the anus (SCCA) is a rare disease often diagnosed at a localised stage. For locally advanced recurrence or metastatic disease, DCF (docetaxel, cisplatin, 5-fluorouracil) demonstrated high efficacy and became one of the standard regimens. However, there is no standard of care in the second line. PATIENTS AND METHODS In the Epitopes-HPV01 and Epitopes-HPV02 prospective trials, 115 patients with advanced SCCA were treated with a DCF regimen in the first line. In these studies, second-line data were registered per protocol. RESULTS After a median follow-up of >40 months, at progression, 73 patients received a second-line (L2) treatment. In this L2 population, median overall survival (mOS) was 13.5 months (95%CI 9.4-19.8), and median progression-free survival (mPFS) was 5.7 months (3.4-7.3) in L2. Fourteen patients presented an oligometastatic progression and were treated with an ablative treatment (surgery or radiotherapy); mOS was 48.3 months (NE-NE), and mPFS was 31.3 months (23.2-NE). Fifty-nine patients received a systemic treatment (chemotherapy or immunotherapy); mOS was 11 months (8.4-15.4) and mPFS was 4.9 months (3.3-7). The most frequent chemotherapy regimens were the reintroduction of DCF, paclitaxel, FOLFIRI and mitomycin plus fluoropyrimidine. No significant difference was observed between regimens (p = 0.26). Six patients received anti-PD1/L1-based immunotherapy. CONCLUSION Second-line treatments are effective in patients with SCCA. Ablative treatment is feasible and is probably the best option for patients with oligometastatic progression. If this is not possible, systemic therapy by an anti-PD1/L1 immunotherapy or chemotherapy can be recommended. Reintroduction of DCF, paclitaxel, FOLFIRI or mitomycin-C plus fluoropyrimidine are possible options.
Collapse
|
105
|
Immunotherapy in Gastrointestinal Malignancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:259-272. [PMID: 34972968 DOI: 10.1007/978-3-030-79308-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Gastrointestinal (GI) cancers represent a heterogeneous group of malignancies, each with a unique tumor biology that in turn affects response to treatment and subsequent prognosis. The interplay between tumor cells and the local immune microenvironment also varies within each GI malignancy and can portend prognosis and response to therapy. Treatment with immune checkpoint inhibitors has changed the treatment landscape of various solid tumors including (but not limited to) renal cell carcinoma, melanoma, and lung cancer. Advances in the understanding between the interplay between the immune system and tumors cells have led to the integration of immunotherapy as standard of care in various GI malignancies. For example, immunotherapy is now a mainstay of treatment for tumors harboring defects in DNA mismatch repair proteins and tumors harboring a high mutational load, regardless of primary site of origin. Data from recent clinical trials have led to the integration of immunotherapy as standard of care for a subset of gastroesophageal cancers and hepatocellular carcinoma. Here, we outline the current landscape of immunotherapy in GI malignancies and highlight ongoing clinical trials that will likely help to further our understanding of how and when to integrate immunotherapy into the treatment of various GI malignancies.
Collapse
|
106
|
Salcedo M, Vinaixa C, Javle M, Trapero-Marugán M, Bustamante J, Line PD. Evaluation and Management of Liver Transplant Candidates With Prior Nonhepatic Cancer: Guidelines From the ILTS/SETH Consensus Conference. Transplantation 2022; 106:e3-e11. [PMID: 34905758 DOI: 10.1097/tp.0000000000003997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Liver transplant in patients with prior nonhepatic cancer is a matter of concern, needing further research, development, and consensus guidelines. This International Liver Transplantation Society/Sociedad Española De Trasplante Hepático consensus conference document focuses on the role of liver transplantation in patients with a prior history of nonhepatic cancer. This document addresses (1) the evaluation of transplant candidates with prior cancers based on the assessment of prognosis, the natural history of individual cancers, and the emerging role for circulating DNA and minimal residual disease in these patients; (2) the impact of prior treatments, including immunotherapy for prior malignancies; and (3) the surveillance of posttransplant cancer recurrence. The consensus statement is based on previously published guidelines, as well as a review of the current, relevant, published literature.
Collapse
Affiliation(s)
- Magdalena Salcedo
- Hepatology and Liver Transplantation Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Carmen Vinaixa
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Department of Hepatology and Liver Transplantation, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Milind Javle
- GI Oncology Unit, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - María Trapero-Marugán
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Universitario Puerta de Hierro Majadahonda, Instituto de Investigación Puerta de Hierro Segovia Arana IDIPHISA, Madrid, Spain
| | - Javier Bustamante
- Liver Transplant and Hepatology Unit, Cruces University Hospital, Baracaldo, Vizcaya, Spain
| | - Pål-Dag Line
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
107
|
Targeted fibroblast growth factor receptor (FGFR) inhibition in recurrent, metastatic anal carcinoma: a case report. Clin Colorectal Cancer 2022; 21:272-275. [DOI: 10.1016/j.clcc.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 01/16/2023]
|
108
|
Tian L, Lei A, Tan T, Zhu M, Zhang L, Mou H, Zhang J. Macrophage-Based Combination Therapies as a New Strategy for Cancer Immunotherapy. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:26-43. [PMID: 35224005 DOI: 10.1159/000518664] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cells of the immune system can inhibit tumor growth and progression; however, immune cells can also promote tumor cell growth, survival, and angiogenesis as a result of the immunosuppressive microenvironments. In the last decade, a growing number of new therapeutic strategies focused on reversing the immunosuppressive status of tumor microenvironments (TMEs), to reprogram the TME to be normal, and to further activate the antitumor functions of immune cells. Most of the "hot tumors" are encompassed with M2 macrophages promoting tumor growth, and the accumulation of M2 macrophages into tumor islets leads to poor prognosis in a wide variety of tumors. SUMMARY Therefore, how to uncover more immunosuppressive signals and to reverse the M2 tumor-associated macrophages (TAMs) to M1-type macrophages is essential for reversing the immunosuppressive state. Except for reeducation of TAMs in the cancer immunotherapy, macrophages as central effectors and regulators of the innate immune system have the capacity of phagocytosis and immune modulation in macrophage-based cell therapies. KEY MESSAGES We review the current macrophage-based cell therapies that use genetic engineering to augment macrophage functionalities with antitumor activity for the application of novel genetically engineered immune cell therapeutics. A combination of TAM reeducation and macrophage-based cell strategy may bring us closer to achieving the original goals of curing cancer. In this review, we describe the characteristics, immune status, and tumor immunotherapy strategies of macrophages to provide clues and evidences for future macrophage-based immune cell therapies.
Collapse
Affiliation(s)
- Lin Tian
- Department of Basic Medical Sciences, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Anhua Lei
- Department of Basic Medical Sciences, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Tianyu Tan
- Department of Basic Medical Sciences, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Mengmeng Zhu
- Department of Basic Medical Sciences, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Li Zhang
- Department of Basic Medical Sciences, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Haibo Mou
- Department of Medical Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou, China
| | - Jin Zhang
- Department of Basic Medical Sciences, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
| |
Collapse
|
109
|
Enomoto N, Yamada K, Terayama M, Kato D, Yagi S, Wake H, Takemura N, Kiyomatsu T, Kokudo N. Current status of immune checkpoint inhibitor therapy for advanced esophageal squamous cell carcinoma. Glob Health Med 2021; 3:378-385. [PMID: 35036619 PMCID: PMC8692100 DOI: 10.35772/ghm.2020.01112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 06/29/2021] [Indexed: 01/27/2023]
Abstract
Esophageal cancer is the seventh most common cancer, with an estimated 572,000 new cases, and the sixth most common cause of cancer-related deaths in 2018 with 509,000 annual worldwide deaths. Advanced esophageal squamous cell carcinoma (ESCC) is one of devastating tumors with a 5-year survival rate of less than 5% in patients with metastatic disease. Treatment options for patients with advanced ESCC are limited. Current guidelines recommend chemotherapy containing a platinum and a fluoropyrimidine agent as a first-line treatment. Recently, immune checkpoint inhibitors (ICIs), including nivolumab and pembrolizumab, have demonstrated antitumor activity and clinical efficacy in patients with advanced ESCC that is refractory or intolerant to first-line chemotherapy. ICIs are game-changers that not only transformed oncological strategy but also have a wide range of clinical potential in combination with conventional cytotoxic chemotherapy and radiotherapy. There is still an urgent, unmet need for reliable treatment options to conquer this intractable disease.
Collapse
Affiliation(s)
- Naoki Enomoto
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kazuhiko Yamada
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masayoshi Terayama
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Daiki Kato
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shusuke Yagi
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hitomi Wake
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Nobuyuki Takemura
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tomomichi Kiyomatsu
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kokudo
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
110
|
Monsrud AL, Avadhani V, Mosunjac MB, Flowers L, Krishnamurti U. Programmed Death Ligand-1 Expression Is Associated With Poorer Survival in Anal Squamous Cell Carcinoma. Arch Pathol Lab Med 2021; 146:1094-1101. [DOI: 10.5858/arpa.2021-0169-oa] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 11/06/2022]
Abstract
Context.—
Upregulation of programmed death ligand-1 (PD-L1), an immunoregulatory protein, is associated with an adverse outcome in several malignancies. Very few studies have evaluated PD-L1 expression in invasive anal squamous cell carcinoma (ASCC).
Objective.—
To assess PD-L1 expression in patients with ASCC and correlate it with clinicopathologic factors and clinical outcomes.
Design.—
Fifty-one cases of ASCC were immunostained for PD-L1. PD-L1 expression by combined positive score and tumor proportion score was correlated with age, gender, HIV status, HIV viral load, CD4 count, stage, and outcomes. Kaplan-Meier curves for overall survival were plotted and compared using the log-rank test. Cox regression analysis was performed to identify significant prognostic factors (2-tailed P < .05 was considered statistically significant).
Results.—
PD-L1 was positive in 24 of 51 cases (47%) by combined positive score and in 18 of 51 (35%) by tumor proportion score. The median cancer-specific survival and 5-year overall survival were significantly lower in PD-L1+ patients. Age, gender, HIV status, HIV viral load, stage, and cancer progression were not significantly different between the two groups. CD4 count of more than 200/μL was significantly higher in PD-L1+ patients. PD-L1+ status remained statistically significant for worse overall survival on multivariate analysis.
Conclusions.—
PD-L1+ status is an independent adverse prognostic factor for overall survival in ASCC. This study highlights the potential of PD-L1 targeted therapy in better management of ASCC.
Collapse
Affiliation(s)
- Ashley L. Monsrud
- From the Department of Pathology & Laboratory Medicine (Monsrud, Avadhani, Mosunjac, Krishnamurti), Emory University, Atlanta, Georgia
| | - Vaidehi Avadhani
- From the Department of Pathology & Laboratory Medicine (Monsrud, Avadhani, Mosunjac, Krishnamurti), Emory University, Atlanta, Georgia
| | - Marina B. Mosunjac
- From the Department of Pathology & Laboratory Medicine (Monsrud, Avadhani, Mosunjac, Krishnamurti), Emory University, Atlanta, Georgia
| | - Lisa Flowers
- Department of Gynecology & Obstetrics (Flowers), Emory University, Atlanta, Georgia
| | - Uma Krishnamurti
- From the Department of Pathology & Laboratory Medicine (Monsrud, Avadhani, Mosunjac, Krishnamurti), Emory University, Atlanta, Georgia
- Krishnamurti is now with the Department of Pathology at Yale School of Medicine
| |
Collapse
|
111
|
Yang N, Xu L, Wang Q, Chen F, Zhou Y. Construction and validation of a prognostic nomogram for anal squamous cell carcinoma. Cancer Med 2021; 11:392-405. [PMID: 34850581 PMCID: PMC8729044 DOI: 10.1002/cam4.4458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
Background Anal squamous cell carcinoma (ASCC) is the main subtype of anal cancer and has great heterogeneity in prognosis. We aimed to construct a nomogram for predicting their 1‐, 3‐, and 5‐year overall survival (OS) rates. Methods Patients with ASCC, enrolled between January 1, 2010 and December 31, 2017, were identified from the SEER database. They were divided into a training group and a validation group in a ratio of 7:3. Univariate and multivariate Cox analyses were used to identify the prognostic factors for OS. Then a prognostic nomogram was established and validated by Harrell consistency index (C‐index), area under the curve (AUC) of the receiver operating characteristic (ROC) curves, calibration plots, and decision curve analysis (DCA). Results We identified 761 patients in training group and 326 patients in validation group. Four prognostic factors including age, sex, AJCC stage, and radiotherapy were identified and integrated to construct a prognostic nomogram. The C‐index and AUC values proved the model's effectiveness and calibration plots manifested its excellent discrimination. Furthermore, in comparison to the AJCC stage, the C‐index, AUC, and DCA proved the nomogram to be of good predictive value. Finally, we constructed a risk stratification model for dividing patients into low‐risk, medium‐risk, and high‐risk groups, and there were obvious differences in OS. Conclusions A prognostic nomogram was firstly established for predicting the survival probability of ASCC patients and helping clinicians improve their risk management.
Collapse
Affiliation(s)
- Ningning Yang
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lu Xu
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qingqing Wang
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fengxia Chen
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
112
|
Yuan Y, Xie WH, Li RZ, Chang H, Zeng ZF, Gao YH, Wang QX, Xiao WW. Comprehensive treatment experience of anal squamous cell carcinoma from a tertiary cancer center in South China. Cancer Med 2021; 11:117-127. [PMID: 34816622 PMCID: PMC8704146 DOI: 10.1002/cam4.4433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/17/2021] [Accepted: 10/31/2021] [Indexed: 12/24/2022] Open
Abstract
Background Anal squamous cell carcinoma (ASCC) is a rare malignant tumor with increasing incidence. The goal of our study was to analyze the treatment outcome and prognostic factors of ASCC in South China in the past half‐century. Methods This study retrospectively included 59 patients with ASCC admitted from 1975 to 2018 in Sun Yat‐sen University cancer center. The clinical records and follow‐up information of all patients were collected. Survival analysis and univariate and multivariate regression analyses were performed using the “survival” and “survminer” packages of R software. Results In 59 patients, 5 patients had distant metastasis at diagnosis. Among 54 M0 stage patients, 33 patients received chemoradiotherapy (CRT), 19 patients received local surgery, and 2 patients refused curative treatment and received the best supportive treatment (BST). The most common grade 3–4 acute toxicities during treatment were myelosuppression and radiation dermatitis. The median follow‐up time was 32 months. For the whole group, the 3‐year and 5‐year overall survival (OS) rates and disease‐free survival (DFS) were 71.1% and 63.6%, and 73.4% and 69.0%, respectively. Multivariate regression analysis showed that the T3–4 stage was an independent prognostic risk factor for OS, progression‐free survival (PFS), and DFS. And M1 was an independent prognostic risk factor for PFS and DFS. Patients in stage M0 mainly treated with CRT had better local control than those mainly treated with surgery (p = 0.027). For M0 patients, induction chemotherapy combined with CRT tends to prolong OS compared with CRT alone (p = 0.26). The 3‐year colostomy‐free survival for the whole group was 81.1%. Conclusions CRT is recommended as the first choice for the treatment of M0 stage ASCC. Induction chemotherapy may bring better survival benefits for some patients. Patients with ASCC in China seem to have a better local control rate, which suggested different treatment strategies may be needed in China.
Collapse
Affiliation(s)
- Yan Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Wei-Hao Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Rong-Zhen Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Hui Chang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Zhi-Fan Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yuan-Hong Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Qiao-Xuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Wei-Wei Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
113
|
Martin-Romano P, Adam J, Scoazec JY, Gouy S, Levy A, Baldini C, Champiat S, Soria JC, Massard C, Marabelle A, Deutsch E, Hollebecque A. Case Report: Response to Immunotherapy, Can Radiotherapy Be a Troublemaker? Front Immunol 2021; 12:745146. [PMID: 34804027 PMCID: PMC8600981 DOI: 10.3389/fimmu.2021.745146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/20/2021] [Indexed: 01/21/2023] Open
Abstract
Immunotherapy has dramatically changed the treatment landscape for several tumor types. However, the impact of previous radiotherapy (RT) on response to immunotherapy is still unknown. We report the case of a 58-year-old female diagnosed with a squamous anal cell carcinoma previously treated with RT and having a dissociated response to anti-PD1 agent. An extensive analysis of the immune contexture performed on the tissue collected from both previously RT-treated and RT-untreated lesions confirmed differences on immune microenvironment, highlighting the potential impact of radiotherapy on the immune response.
Collapse
Affiliation(s)
- Patricia Martin-Romano
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
| | - Julien Adam
- Department of Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jean-Yves Scoazec
- Department of Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sébastien Gouy
- Department of Surgery, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Capucine Baldini
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
| | - Stéphane Champiat
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
| | - Jean-Charles Soria
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Christophe Massard
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
| | - Aurélien Marabelle
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Antoine Hollebecque
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
114
|
Armstrong SA, Malley R, Wang H, Lenz HJ, Arguello D, El-Deiry WS, Xiu J, Gatalica Z, Hwang JJ, Philip PA, Shields AF, Marshall JL, Salem ME, Weinberg BA. Molecular characterization of squamous cell carcinoma of the anal canal. J Gastrointest Oncol 2021; 12:2423-2437. [PMID: 34790403 DOI: 10.21037/jgo-20-610] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Squamous cell carcinoma of the anal canal (SCCA) is an uncommon malignancy with limited therapeutic options. Nivolumab and pembrolizumab show promising results in patients with SCCA. Human papillomavirus (HPV)-negative tumors are frequently TP53-mutated (TP53-MT) and often resistant to therapy. Methods We present a large molecularly-profiled cohort of SCCA, exploring the underlying biology of SCCA, differences between TP53-wild type (TP53-WT) and TP53-MT tumors, and differences between local and metastatic tumors. SCCA specimens (n=311) underwent multiplatform testing with immunohistochemistry (IHC), in situ hybridization (ISH) and next-generation sequencing (NGS). Tumor mutational burden (TMB) was calculated using only somatic nonsynonymous missense mutations. Chi-square testing was used for comparative analyses. Results The most frequently mutated genes included PIK3CA (28.1%), KMT2D (19.5%), FBXW7 (12%), TP53 (12%) and PTEN (10.8%). The expression of PD-1 was seen in 68.8% and PD-L1 in 40.5% of tumors. High TMB was present in 6.7% of specimens. HER2 IHC was positive in 0.9%, amplification by chromogenic in situ hybridization (CISH) was seen 1.3%, and mutations in ERBB2 were present in 1.8% of tumors. The latter mutation has not been previously described in SCCA. When compared with TP53-WT tumors, TP53-MT tumors had higher rates of CDKN2A, EWSR1, JAK1, FGFR1 and BRAF mutations. PD-1 and PD-L1 expression were similar, and high TMB did not correlate with PD-1 (P=0.50) or PD-L1 (P=0.52) expression. Conclusions Molecular profiling differences between TP53-MT and TP53-WT SCCA indicate different carcinogenic pathways which may influence response to therapy. Low frequency mutations in several druggable genes may provide therapeutic opportunities for patients with SCCA.
Collapse
Affiliation(s)
- Samantha A Armstrong
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Rita Malley
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Hongkun Wang
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, Los Angeles, CA, USA
| | | | | | | | | | - Jimmy J Hwang
- Department of Hematology/Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Philip A Philip
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - John L Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Mohamed E Salem
- Department of Hematology/Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
115
|
Moniz CMV, Riechelmann RP, Oliveira SCR, Bariani GM, Rivelli TG, Ortega C, Pereira AAL, Meireles SI, Franco R, Chen A, Bonadio RC, Nahas C, Sabbaga J, Coudry RA, Braghiroli MI, Hoff PM. A Prospective Cohort Study of Biomarkers in Squamous Cell Carcinoma of the Anal Canal (SCCAC) and their Influence on Treatment Outcomes. J Cancer 2021; 12:7018-7025. [PMID: 34729104 PMCID: PMC8558650 DOI: 10.7150/jca.57678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/05/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Although Chemoradiation (CRT) is the curative treatment for SCCAC, many patients present primary resistance. Since it is a rare tumor, response predictors remain unknown. Methods: We performed a prospective cohort study to evaluate biomarkers associated with CRT response, progression-free survival (PFS), and overall survival (OS). The primary endpoint was response at 6 months (m). Tumor DNA and HPV were analyzed by next-generation sequencing, while KI-67 and PD-L1 by immunohistochemistry in tumor tissue. Results: Seventy-eight patients were recruited between October/2011 and December/2015, and 75 were response evaluable. The median age was 57 years, 65% (n=49) were stage III and 12% (n=9) were HIV positive (HIV+). At 6m, 62.7% (n=47) presented CR. On multivariate analyses, stage II patients were 4.7 more likely to achieve response than stage III (OR, 4.70; 95%CI, 1.36-16.30; p=0.015). HIV+ was associated with a worse response (OR, 5.72; 95%CI, 2.5-13.0; p<0.001). 5-year PFS and OS rates were 63.3% and 76.4%, respectively, with a median follow up of 66m. On multivariate analyses, older age (HR 1.06, p=0.022, 95%IC 1.01-1.11) and absence of CR at 6m (HR 3.36, p=0.007, 95%IC 1.39-8.09) were associated with inferior OS. The 5-year OS rate was 62.5% in HIV+ group compared to 78% among HIV- pts, although this difference was not statistically significant (p=0.4). PIK3CA, MET and TP53 mutations, HPV, Ki-67 expression, and PD-L1 expression, were not associated with PFS and OS. Conclusions: Clinical stage III and HIV+ were associated with worse response to CRT at 6m. The absence of CR was the main factor associated with poor 5-year OS.
Collapse
Affiliation(s)
- Camila Motta Venchiarutti Moniz
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| | | | | | - Giovanni Mendonça Bariani
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Thomas Giollo Rivelli
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Cintia Ortega
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | | | - Rejane Franco
- Universidade Federal do Paraná - Hospital de Clínicas, Curitiba, PR, Brasil
| | - Andre Chen
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Renata Colombo Bonadio
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Caio Nahas
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Jorge Sabbaga
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Maria Ignez Braghiroli
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| | - Paulo Marcelo Hoff
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| |
Collapse
|
116
|
Lonardi S, Prete AA, Morano F, Messina M, Formica V, Corsi DC, Orciuolo C, Frassineti GL, Zampino MG, Casagrande M, Masi G, Ronzoni M, Scartozzi M, Buonadonna A, Mosconi S, Ratti M, Sartore-Bianchi A, Tamburini E, Prisciandaro M, Bergamo F, Spada M, Corallo S, Vettore V, Loupakis F, Fassan M, Del Bianco P, Zagonel V, Pietrantonio F. Randomized phase II trial of avelumab alone or in combination with cetuximab for patients with previously treated, locally advanced, or metastatic squamous cell anal carcinoma: the CARACAS study. J Immunother Cancer 2021; 9:e002996. [PMID: 34815354 PMCID: PMC8611452 DOI: 10.1136/jitc-2021-002996] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND No standard therapies beyond first line are established for advanced squamous cell anal carcinoma (aSCAC). Earlier preliminary data suggest activity of epidermal growth factor receptor (EGFR) inhibition and programmed cell death ligand (PD-(L))1 blockade in patients with previously treated disease. Aim of this study was to explore activity and safety of avelumab with/without cetuximab in patients with aSCAC. METHODS In this open-label, non-comparative, 'pick the winner', multicenter randomized phase II trial (NCT03944252), patients with aSCAC progressing after one or more lines of treatment were randomized 1:1 to the anti-PD-L1 agent avelumab alone (arm A) or combined with cetuximab (arm B). Overall response rate (ORR) was the primary endpoint. With one-sided α error set at 0.05 and power of 80%, at least 4 responses out of 27 patients per arm had to be observed to declare the study positive. Secondary endpoints were progression free survival (PFS), overall survival (OS), and safety. RESULTS Thirty patients per arm were enrolled. Three patients in arm A and five in arm B achieved partial response: primary endpoint was reached in combination arm. ORR was 10% (95% CI 2.1 to 26.5) and 17% (95% CI 5.6 to 34.7) in arms A and B; disease control rate was 50% (95% CI 31.3 to 68.7) in arm A and 57 (95% CI 37.4-74.5) in arm B. At a median follow-up of 26.7 months (IQR 26.5-26.9), median PFS was 2.0 months (95% CI 1.8 to 4.0) in arm A and 3.9 (95% CI 2.1 to 5.6) in arm B. Median OS was 13.9 months (95% CI 7.7 to 19.4) in arm A and 7.8 (95% CI 6.2 to 11.2) in arm B. Acceptable safety profile was observed in both arms. CONCLUSIONS CARACAS study met its primary endpoint in arm B, documenting promising activity of dual EGFR and PD-L1 blockade in aSCAC.
Collapse
Affiliation(s)
- Sara Lonardi
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology 1, veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Alessandra Anna Prete
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology 1, veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Marco Messina
- Department of Oncology, Fondazione Istituto G. Giglio, Cefalù, Italy
| | | | | | - Corrado Orciuolo
- Oncology Unit, Department of Radiology, Oncology and Human Pathology, Sapienza University of Roma, Rome, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori Dino Amadori (IRST), Meldola, Italy
| | - Maria Giulia Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology-IRCCS, Milan, Italy
| | - Mariaelena Casagrande
- Department of Oncology, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Gianluca Masi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Division of Medical Oncology, Pisa University Hospital, Pisa, Italy
| | - Monica Ronzoni
- Oncologia Medica, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Cagliari, Italy
| | - Angela Buonadonna
- Unit of Medical Oncology and Cancer Prevention, Centro di Riferimento Oncologico di Aviano IRCCS, Aviano, Italy
| | - Stefania Mosconi
- Department of Medical Oncology, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Emiliano Tamburini
- Department of Oncology and Palliative Care, Cardinale G Panico, Tricase City Hospital, Tricase, Italy
| | - Michele Prisciandaro
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Francesca Bergamo
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology 1, veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Salvatore Corallo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Valentina Vettore
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology 1, veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Fotios Loupakis
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology 1, veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Pathology Unit, University of Padua, Padua, Veneto, Italy, University of Padova, Padova, Veneto, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Veneto, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Vittorina Zagonel
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology 1, veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
117
|
Guerra GR, Kong JC, Millen RM, Read M, Liu DS, Roth S, Sampurno S, Sia J, Bernardi MP, Chittleborough TJ, Behrenbruch CC, Teh J, Xu H, Haynes NM, Yu J, Lupat R, Hawkes D, Di Costanzo N, Tothill RW, Mitchell C, Ngan SY, Heriot AG, Ramsay RG, Phillips WA. Molecular and genomic characterisation of a panel of human anal cancer cell lines. Cell Death Dis 2021; 12:959. [PMID: 34663790 PMCID: PMC8523722 DOI: 10.1038/s41419-021-04141-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022]
Abstract
Anal cancer is a rare disease that has doubled in incidence over the last four decades. Current treatment and survival of patients with this disease has not changed substantially over this period of time, due, in part, to a paucity of preclinical models to assess new therapeutic options. To address this hiatus, we set-out to establish, validate and characterise a panel of human anal squamous cell carcinoma (ASCC) cell lines by employing an explant technique using fresh human ASCC tumour tissue. The panel of five human ASCC cell lines were validated to confirm their origin, squamous features and tumourigenicity, followed by molecular and genomic (whole-exome sequencing) characterisation. This panel recapitulates the genetic and molecular characteristics previously described in ASCC including phosphoinositide-3-kinase (PI3K) mutations in three of the human papillomavirus (HPV) positive lines and TP53 mutations in the HPV negative line. The cell lines demonstrate the ability to form tumouroids and retain their tumourigenic potential upon xenotransplantation, with varied inducible expression of major histocompatibility complex class I (MHC class I) and Programmed cell death ligand 1 (PD-L1). We observed differential responses to standard chemotherapy, radiotherapy and a PI3K specific molecular targeted agent in vitro, which correlated with the clinical response of the patient tumours from which they were derived. We anticipate this novel panel of human ASCC cell lines will form a valuable resource for future studies into the biology and therapeutics of this rare disease.
Collapse
Affiliation(s)
- Glen R Guerra
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Joseph C Kong
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rosemary M Millen
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew Read
- Department of Surgery, St Vincent's Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - David S Liu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- UGI Surgery Unit, Austin Hospital, 145 Studley Road, Heidelberg, Victoria, 3084, Australia
| | - Sara Roth
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Shienny Sampurno
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Joseph Sia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Maria-Pia Bernardi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Timothy J Chittleborough
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Corina C Behrenbruch
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jiasian Teh
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Huiling Xu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Nicole M Haynes
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jiaan Yu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Richard Lupat
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - David Hawkes
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
- VCS Foundation, Carlton, VIC, 3053, Australia
- Department of Pathology, University of Malaya, Kuala Lumpur, Malaysia
| | - Natasha Di Costanzo
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Richard W Tothill
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Centre for Cancer Research, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Samuel Y Ngan
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Alexander G Heriot
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Surgery, St Vincent's Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Robert G Ramsay
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wayne A Phillips
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Department of Surgery, St Vincent's Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
118
|
Anti-PD1 antibody enhances the anti-tumor efficacy of MUC1-MBP fusion protein vaccine via increasing Th1, Tc1 activity and decreasing the proportion of MDSC in the B16-MUC1 melanoma mouse model. Int Immunopharmacol 2021; 101:108173. [PMID: 34607233 DOI: 10.1016/j.intimp.2021.108173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022]
Abstract
In previous studies, we have obtained a notable anti-tumor efficacy of the recombinant MUC1-MBP vaccine in the process of mouse B16-MUC1 melanoma treatment. However, the tumor cannot be eliminated completely. We found that the tumor inhibition rate decreased from 81.67% (five immunizations) to 43.67% (eight immunizations) after more than five immunizations, indicating persistent vaccine stimulation may activate immunosuppressive factors. In the present study, we revealed that programmed cell death 1 (PD1), an inhibitory molecule suppressing T cell function, expressed on splenic and tumor-infiltrating T cells were up-regulated by the vaccine. Therefore, to optimize the anti-tumor efficacy of the vaccine, we employed combination immunotherapy with MUC1-MBP vaccine and αPD1 (anti-PD1 antibody). Results showed that combination immunotherapy induced a more remarkable anti-tumor efficacy, the tumor clearance being increased to 80% from 20% which obtain by MUC1-MBP vaccine immunizations. To investigate the possible underlying mechanism, IFN-γ secretion and cytotoxic T lymphocyte (CTL) cytotoxicity were measured by enzyme-linked immunosorbent assay (ELISA) and xCELLigence real-time cell analyzer (RTCA) respectively. T cell subsets and immunosuppressive cells in the mouse spleen and tumor microenvironment were analyzed by FACS. Results showed that the proportion of splenic CD8+T cells and tumor infiltration was increased and the activity of CTL killing, T helper 1 (Th1), Type 1 CD8+T (Tc1) was enhanced, indicating that the anti-tumor efficacy enhanced by combination immunotherapy was mainly through boosting CD8+T cells mediated anti-tumor cellular immunity. Additionally, combination immunotherapy significantly decreased the splenic and tumor-infiltrating myeloid derived suppressor cells (MDSCs). These results demonstrated that combination immunotherapy with MUC1-MBP vaccine and αPD1 was capable to invoke a more potent anti-tumor immune response and provide a foundation for further research.
Collapse
|
119
|
Liu L, Shi W, Xiao X, Wu X, Hu H, Yuan S, Liu K, Liu Z. BCG immunotherapy inhibits cancer progression by promoting the M1 macrophage differentiation of THP‑1 cells via the Rb/E2F1 pathway in cervical carcinoma. Oncol Rep 2021; 46:245. [PMID: 34581419 PMCID: PMC8493057 DOI: 10.3892/or.2021.8196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 07/19/2021] [Indexed: 01/01/2023] Open
Abstract
Bacillus Calmette-Guérin (BCG) immunotherapy increases macrophage polarization toward M1-type macrophages. In the present study, to identify the M1/M2 marker genes in the carcinogenesis and progression of cervical cancer, the microarray datasets GSE9750 and GSE7803 were downloaded from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and the University of California Santa Cruz (UCSC) Xena browser. Survival analysis revealed that M1 markers (IL-12) were involved in anti-tumour progression, and M2 markers (IL-10) were involved in the carcinogenesis and invasion of cervical cancer. The expression of M1 markers (IL-12, inducible nitric oxide synthase and CD80) and M2 markers (IL-10 and arginase) was examined to determine whether BCG affects the polarization of macrophages and to elucidate the underlying mechanisms. The results revealed that BCG promoted macrophage polarization towards the M1 phenotype and enhanced the transition of M2 to M1 macrophages. The results also revealed that polarized M1 macrophages induced by BCG decreased the protein expression of phosphorylated (p-)retinoblastoma (Rb)/E2F transcription factor 1 (E2F1), inhibited the proliferation and promoted the apoptosis of HeLa cells. On the whole, these results demonstrated that BCG promoted the anti-tumour progression of M1 macrophages and inhibited the pro-tumour activation of M2 macrophages via the Rb/E2F1 signalling pathway in HeLa cells. This suggests the possibility of a direct translation of this combination strategy to clinical practice for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Limin Liu
- Department of Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Wenjuan Shi
- Department of Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Xiao Xiao
- Department of Obstetrics, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Xuemei Wu
- Department of Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Haiyan Hu
- Department of Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Shixin Yuan
- Institute of Maternal and Child Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Kai Liu
- Department of Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Zhihua Liu
- Department of Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| |
Collapse
|
120
|
Strauss J, Gatti-Mays ME, Cho BC, Hill A, Salas S, McClay E, Redman JM, Sater HA, Donahue RN, Jochems C, Lamping E, Burmeister A, Marté JL, Cordes LM, Bilusic M, Karzai F, Ojalvo LS, Jehl G, Rolfe PA, Hinrichs CS, Madan RA, Schlom J, Gulley JL. Bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in patients with human papillomavirus-associated malignancies. J Immunother Cancer 2021; 8:jitc-2020-001395. [PMID: 33323462 PMCID: PMC7745517 DOI: 10.1136/jitc-2020-001395] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Bintrafusp alfa is a first-in-class bifunctional fusion protein composed of the extracellular domain of transforming growth factor (TGF)-βRII (a TGF-β 'trap') fused to a human IgG1 mAb blocking programmed cell death ligand 1. This is the largest analysis of patients with advanced, pretreated human papillomavirus (HPV)-associated malignancies treated with bintrafusp alfa. METHODS In these phase 1 (NCT02517398) and phase 2 trials (NCT03427411), 59 patients with advanced, pretreated, checkpoint inhibitor-naive HPV-associated cancers received bintrafusp alfa intravenously every 2 weeks until progressive disease, unacceptable toxicity, or withdrawal. Primary endpoint was best overall response per Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1; other endpoints included safety. RESULTS As of April 17, 2019 (phase 1), and October 4, 2019 (phase 2), the confirmed objective response rate per RECIST V.1.1 in the checkpoint inhibitor-naive, full-analysis population was 30.5% (95% CI, 19.2% to 43.9%; five complete responses); eight patients had stable disease (disease control rate, 44.1% (95% CI, 31.2% to 57.6%)). In addition, three patients experienced a delayed partial response after initial disease progression, for a total clinical response rate of 35.6% (95% CI, 23.6% to 49.1%). An additional patient with vulvar cancer had an unconfirmed response. Forty-nine patients (83.1%) experienced treatment-related adverse events, which were grade 3/4 in 16 patients (27.1%). No treatment-related deaths occurred. CONCLUSION Bintrafusp alfa showed clinical activity and manageable safety and is a promising treatment in HPV-associated cancers. These findings support further investigation of bintrafusp alfa in patients with advanced, pretreated HPV-associated cancers.
Collapse
Affiliation(s)
- Julius Strauss
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Margaret E Gatti-Mays
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Byoung Chul Cho
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Andrew Hill
- Tasman Oncology Research Ltd, Southport, Queensland, Australia
| | - Sébastien Salas
- CEPCM Assistance Publique des Hôpitaux de Marseille; Aix-Marseille Université, Marseille, France
| | - Edward McClay
- California Cancer Associates for Research and Excellence, Encinitas, California, USA
| | - Jason M Redman
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Houssein A Sater
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth Lamping
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrea Burmeister
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.,Leidos Biomedical Research, Frederick, Maryland, USA
| | - Jennifer L Marté
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa M Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Laureen S Ojalvo
- EMD Serono Research & Development Institute, Inc, Billerica, Massachusetts, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | | | - P Alexander Rolfe
- EMD Serono Research & Development Institute, Inc, Billerica, Massachusetts, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Christian S Hinrichs
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
121
|
Guren MG, Sebag-Montefiore D, Franco P, Johnsson A, Segelov E, Deutsch E, Rao S, Spindler KLG, Arnold D. Treatment of Squamous Cell Carcinoma of the Anus, Unresolved Areas and Future Perspectives for Research: Perspectives of Research Needs in Anal Cancer. Clin Colorectal Cancer 2021; 20:279-287. [PMID: 34645589 DOI: 10.1016/j.clcc.2021.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/30/2021] [Accepted: 09/09/2021] [Indexed: 01/12/2023]
Abstract
Anal cancer is a relatively rare, mostly HPV-related cancer. The curative treatment consists of concurrent chemoradiation delivered with modern radiotherapy techniques. The prognosis for most patients with early localized disease is very favourable; however patients with locally advanced disease and/or HPV negative tumours are at higher risk of locoregional and distant treatment failure. Tailored approaches are presently being investigated to determine the most suitable regimen in terms of radiotherapy dose prescription, target volume selection, normal tissue avoidance, and combination therapy. Metastatic anal cancer is treated with chemotherapy aiming at prolonged survival. The role of immune therapy in the clinical setting is being investigated. There is little knowledge on the biology of anal cancer, and an urgent need for more clinical and translational research dedicated to this disease. In this article, the evidence-base for the current treatment is briefly reviewed, and perspectives on future research needs are high-lighted.
Collapse
Affiliation(s)
| | | | - Pierfrancesco Franco
- Department of Translational Medicine, University of Eastern Piedmont and Department of Radiation Oncology, AOU ''Maggiore della Carità,'' Novara, Italy
| | - Anders Johnsson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Eva Segelov
- School of Clinical Sciences, Faculty of Medicine, Monash University, Clayton, Australia and Department of Oncology, Monash Health Clayton, Australia
| | | | - Sheela Rao
- GI Unit, Royal Marsden Hospital, London, UK
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| |
Collapse
|
122
|
Rao S, Guren MG, Khan K, Brown G, Renehan AG, Steigen SE, Deutsch E, Martinelli E, Arnold D. Anal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up ☆. Ann Oncol 2021; 32:1087-1100. [PMID: 34175386 DOI: 10.1016/j.annonc.2021.06.015] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- S Rao
- GI Unit, Royal Marsden Hospital, London, UK
| | - M G Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - K Khan
- University College London Hospitals NHS Foundation Trust/UCL Cancer Institute, London, UK; Royal Marsden Hospital, London, UK
| | - G Brown
- Department of Radiology, Royal Marsden NHS Foundation Trust, London, UK
| | - A G Renehan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Christie NHS Foundation Trust, Manchester, UK
| | - S E Steigen
- University Hospital of North Norway, Tromsø, Norway
| | - E Deutsch
- INSERM 1030, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - E Martinelli
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - D Arnold
- Department of Hematology, Oncology, Palliative Care Medicine and Rheumatology, Asklepios Hospital Altona, Hamburg, Germany
| |
Collapse
|
123
|
Abstract
Owing to the presence of known tumor-specific viral antigens, human papillomavirus (HPV)-associated cancers are well suited for treatment with immunotherapy designed to unleash, amplify or replace the T cell arm of the adaptive immune system. Immune checkpoint blockade designed to unleash existing T cell immunity is currently Food and Drug Administration approved for certain HPV-associated cancers. More specific immunotherapies such as therapeutic vaccines and T cell receptor-engineered cellular therapy are currently in clinical development. Such therapies may offer more specific immune activation against viral tumor antigens and decrease the risk of immune-related adverse events. Current and planned clinical study of these treatments will determine their utility in the treatment of patients with newly diagnosed advanced stage or relapsed HPV-associated cancer.
Collapse
Affiliation(s)
- Maxwell Y Lee
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
124
|
Turchan WT, Liauw SL. Chemoradiation for Anal Cancer: Clinical Outcomes and Strategies to Optimize the Therapeutic Ratio According to HPV Status. Semin Radiat Oncol 2021; 31:349-360. [PMID: 34455990 DOI: 10.1016/j.semradonc.2021.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The incidence of anal cancer in the United States has increased in recent years, primarily related to the increasing incidence of HPV-associated anal squamous cell carcinoma, which is estimated to represent 80%-95% of anal cancers. Similar to head and neck cancer, HPV association has been demonstrated to be a strong positive prognostic factor in patients with anal cancer. Encouraging results from a number of studies investigating treatment de-escalation for HPV-associated oropharyngeal cancer support the notion that similar attempts may be feasible in HPV-associated anal cancer; however, the data to support this hypothesis are currently lacking. Studies are needed to determine how, if at all, HPV status should impact the management of patients with anal cancer. This review summarizes the relationship between HPV association and outcomes for patients with anal cancer, and how HPV status may impact the treatment of patients with anal cancer going forward.
Collapse
Affiliation(s)
| | - Stanley L Liauw
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL.
| |
Collapse
|
125
|
Castelli V, Lombardi A, Palomba E, Bozzi G, Ungaro R, Alagna L, Mangioni D, Muscatello A, Bandera A, Gori A. Immune Checkpoint Inhibitors in People Living with HIV/AIDS: Facts and Controversies. Cells 2021; 10:2227. [PMID: 34571876 PMCID: PMC8467545 DOI: 10.3390/cells10092227] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are reshaping the landscape of cancer treatment, redefining the prognosis of several tumors. They act by restoring the cytotoxic activity of tumor-specific T lymphocytes that are in a condition of immune exhaustion. The same condition has been widely described in chronic HIV infection. In this review, we dissect the role of ICIs in people living with HIV/AIDS (PLWHIV). First, we provide an overview of the immunologic scenario. Second, we discuss the possible use of ICIs as adjuvant treatment of HIV to achieve elimination of the viral reservoir. Third, we examine the influence of HIV infection on ICI safety and effectiveness. Finally, we describe how the administration of ICIs impacts opportunistic infections.
Collapse
Affiliation(s)
- Valeria Castelli
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Andrea Lombardi
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
| | - Emanuele Palomba
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Giorgio Bozzi
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Riccardo Ungaro
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Laura Alagna
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Davide Mangioni
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Antonio Muscatello
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, 20122 Milano, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (V.C.); (E.P.); (G.B.); (R.U.); (L.A.); (D.M.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milano, 20122 Milano, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, 20122 Milano, Italy
| |
Collapse
|
126
|
Spehner L, Boustani J, Cabel L, Doyen J, Vienot A, Borg C, Kim S. Present and Future Research on Anal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:3895. [PMID: 34359795 PMCID: PMC8345786 DOI: 10.3390/cancers13153895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Squamous cell carcinoma of the anus is an orphan disease, and after more than three decades of no substantial advances in disease knowledge and treatment, it is finally gaining momentum with the arrival of a taxane-based chemotherapy and immunotherapy. Currently, about 20 combination clinical trials with an anti-PD1/L1 are ongoing in localized and advanced stages, in association with radiotherapy, chemotherapy, tumor vaccines, anti-CTLA4, anti-EGFR, or antiangiogenic molecules. Moreover, a new biomarker with high sensitivity and specificity such as HPV circulating tumor DNA (HPV ctDNA) by liquid biopsy, is improving not only the prognostic measurement but also the treatment strategy guidance for this disease. Finally, better understanding of potential targets is reshaping the present and future clinical research in this unique, HPV genotype-16-related disease in the great majority of patients.
Collapse
Affiliation(s)
- Laurie Spehner
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
| | - Jihane Boustani
- Department of Radiotherapy, University Hospital of Besançon, 25030 Besançon, France;
| | - Luc Cabel
- Department of Medical Oncology, Curie Institute, 75005 Paris, France;
| | - Jérôme Doyen
- Department of Medical Oncology, Centre Antoine-Lacassagne, 06189 Nice, France;
| | - Angélique Vienot
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, 25030 Besançon, France
| | - Christophe Borg
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, 25030 Besançon, France
| | - Stefano Kim
- Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique Research Unit INSERM UMR1098, University of Bourgogne Franche-Comté, 25020 Besançon, France; (L.S.); (A.V.); (C.B.)
- Department of Medical Oncology, University Hospital of Besançon, 25030 Besançon, France
- Clinical Investigational Center, INSERM CIC-1431, Centre Hospitalier Universitaire de Besançon, 25030 Besançon, France
- Department of Oncology and Radiotherapy, Nord Franche Comté Hospital, 25209 Montbéliard, France
| |
Collapse
|
127
|
Shamseddine AA, Burman B, Lee NY, Zamarin D, Riaz N. Tumor Immunity and Immunotherapy for HPV-Related Cancers. Cancer Discov 2021; 11:1896-1912. [PMID: 33990345 PMCID: PMC8338882 DOI: 10.1158/2159-8290.cd-20-1760] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Human papillomavirus (HPV) infection drives tumorigenesis in the majority of cervical, oropharyngeal, anal, and vulvar cancers. Genetic and epidemiologic evidence has highlighted the role of immunosuppression in the oncogenesis of HPV-related malignancies. Here we review how HPV modulates the immune microenvironment and subsequent therapeutic implications. We describe the landscape of immunotherapies for these cancers with a focus on findings from early-phase studies exploring antigen-specific treatments, and discuss future directions. Although responses across these studies have been modest to date, a deeper understanding of HPV-related tumor biology and immunology may prove instrumental for the development of more efficacious immunotherapeutic approaches. SIGNIFICANCE: HPV modulates the microenvironment to create a protumorigenic state of immune suppression and evasion. Our understanding of these mechanisms has led to the development of immunomodulatory treatments that have shown early clinical promise in patients with HPV-related malignancies. This review summarizes our current understanding of the interactions of HPV and its microenvironment and provides insight into the progress and challenges of developing immunotherapies for HPV-related malignancies.
Collapse
Affiliation(s)
- Achraf A Shamseddine
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bharat Burman
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dmitriy Zamarin
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
128
|
Martini G, Arrichiello G, Borrelli C, Poliero L, Martinelli E. How I treat anal squamous cell carcinoma. ESMO Open 2021; 4:e000711. [PMID: 32883674 PMCID: PMC7473616 DOI: 10.1136/esmoopen-2020-000711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022] Open
Abstract
Squamous cell carcinoma of the anus (SCCA) is a rising health issue, strongly related to other relevant medical conditions such as (HIV) and human papillomavirus (HPV) infection. Correct assessment of patients with SCCA requires a multidisciplinary evaluation and adequate follow-up. Accurate local and systemic staging, as well as risk evaluation, are essential to optimal treatment planning. Early stage tumours can be definitively treated with a combination of chemotherapy and radiotherapy, while salvage surgery is usually reserved for patients who develop local recurrence. Distant recurrence and de novo metastatic disease are associated with poorer prognosis and require palliative systemic chemotherapy, with different single agent and combination options available. Finally, recent discoveries on the carcinogenesis of SCCA have allowed the development of innovative treatment options, the most promising being immune checkpoint inhibitors. The limited systemic treatments for SCCA and low incidence of the disease, together with insufficient data from clinical research could explain the poor outcomes of these patients, which should therefore be managed in high volume centres and enrolled in clinical trials whenever possible. This article summarises the main strategies for treating patients with SCCA.
Collapse
Affiliation(s)
- Giulia Martini
- Medical Oncology, Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy
| | - Gianluca Arrichiello
- Medical Oncology, Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy
| | - Carola Borrelli
- Medical Oncology, Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy
| | - Luca Poliero
- Medical Oncology, Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy
| | - Erika Martinelli
- Medical Oncology, Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy.
| |
Collapse
|
129
|
Giunta EF, Bregni G, Hendlisz A, Sclafani F. Anal squamous cell carcinoma: standards of care, new data and ongoing clinical trials. Curr Opin Oncol 2021; 33:372-377. [PMID: 33882526 DOI: 10.1097/cco.0000000000000748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To summarize current standards of care, discuss results of recent studies and present ongoing clinical trials for anal squamous cell carcinoma (ASCC). RECENT FINDINGS Over the last year, no practice changing studies have been reported in the setting of localised ASCC. A number of retrospective analyses, however, have provided practice-informing data, such as those confirming the negative impact of low compliance to chemoradiotherapy (CRT) on patient outcomes. In contrast, and for the first time, randomized evidence has become available to inform the management of advanced tumours. The InterAACT trial represents a key milestone in the evidence-building process for this disease, establishing carboplatin plus paclitaxel as a new standard of care for treatment-naïve advanced ASCC patients. Furthermore, more data have accumulated about the value of triplet chemotherapy in the first-line setting and of immune checkpoint inhibitors (either as single agents or in combination with other agents) in the refractory setting. SUMMARY Recent findings have the potential to improve the treatment quality standards and overall outcome of patients with either localised or advanced ASCC. Results from ongoing clinical trials will hopefully provide useful insights into the management of this disease and further shape current treatment paradigms.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Medical Oncology Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Giacomo Bregni
- Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alain Hendlisz
- Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Francesco Sclafani
- Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
130
|
Slørdahl KS, Klotz D, Olsen JÅ, Skovlund E, Undseth C, Abildgaard HL, Brændengen M, Nesbakken A, Larsen SG, Hanekamp BA, Holmboe L, Tvedt R, Sveen A, Lothe RA, Malinen E, Kaasa S, Guren MG. Treatment outcomes and prognostic factors after chemoradiotherapy for anal cancer. Acta Oncol 2021; 60:921-930. [PMID: 33966592 DOI: 10.1080/0284186x.2021.1918763] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Squamous cell carcinoma of the anus (SCCA) is a rare malignancy with rising incidence, associated with human papilloma virus (HPV). Chemoradiotherapy (CRT) is the preferred treatment. The purpose was to investigate treatment failure, survival and prognostic factors after CRT. MATERIAL AND METHODS In this prospective observational study from a large regional centre, 141 patients were included from 2013 to 2017, and 132 were eligible for analysis. The main inclusion criteria were SCCA, planned radiotherapy, and performance status (ECOG) ≤2. Patient characteristics, disease stage, treatment, and treatment response were prospectively registered. Disease-free survival (DFS), overall survival (OS), and locoregional treatment failure after CRT were analysed. Hazard ratios (HRs) were estimated with Cox`s proportional hazards model. RESULTS Median follow-up was 54 (range 6-71) months. Eighteen patients (14%) had treatment failures after CRT; of these 10 (8%) had residual tumour, and 8 (6%) relapse as first failure. The first treatment failure was locoregional (11 patients), distant (5 patients), and both (2 patients). Salvage abdomino-perineal resection was performed in 10 patients, 2 had resections of metastases, and 3 both. DFS was 85% at 3 years and 78% at 5 years. OS was 93% at 3 years and 86% at 5 years. In analyses adjusted for age and gender, HPV negative tumours (HR 2.5, p = 0.024), N3 disease (HR 2.6, p = 0.024), and tumour size ≥4 cm (HR 2.4, p = 0.038) were negative prognostic factors for DFS. CONCLUSION State-of-the-art chemoradiotherapy for SCCA resulted in excellent outcomes, and improved survival compared with previous national data, with <15% treatment failures and a 3-year DFS of >80%.
Collapse
Affiliation(s)
- Kathinka S. Slørdahl
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Dagmar Klotz
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Jan-Åge Olsen
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Eva Skovlund
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Morten Brændengen
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Arild Nesbakken
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Stein Gunnar Larsen
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Bettina A. Hanekamp
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Laila Holmboe
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Ragnhild Tvedt
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Anita Sveen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
- Department of Molecular Oncology, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A. Lothe
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
- Department of Molecular Oncology, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Physics, University of Oslo, Oslo, Norway
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
| | - Stein Kaasa
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marianne Grønlie Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
131
|
Intensified Induction Chemotherapy in Locally Advanced Squamous Cell Carcinoma of the Anus-A Population-Based Experience from the Danish Anal Cancer Group. Cancers (Basel) 2021; 13:cancers13133226. [PMID: 34203394 PMCID: PMC8267651 DOI: 10.3390/cancers13133226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary The primary treatment modality for anal cancer is chemoradiotherapy, but patients with locally advanced disease (i.e., large tumors and/or involvement of regional lymph nodes) have a high risk of treatment failure. The use of chemotherapy prior to radiotherapy (induction chemotherapy) can potentially shrink the tumor and/or eradicate small cancer cells with metastatic potential, with a chance of a better outcome. With this paper, the authors present 20 years of nationwide experience with intensified induction chemotherapy in the treatment of locally advanced anal cancer, which indicates a role for further investigation in the most advanced cases. Abstract Locally advanced squamous cell carcinoma of the anus (LASCCA) has a poor prognosis with a high risk of treatment failure calling for intensified therapy. We present the long-term follow-up of a nationwide cohort of LASCCA treated with intensified induction chemotherapy (ICT). The study included patients with LASCCA (T3-4N0 or T1-4N+) treated with at least one cycle of ICT (cisplatin, ifosfamide, leucoverin, and 5-flourouracil) between 1998–2018. Data were retrospectively collected from medical records, and statistics were performed in STATA 16.1. In total, 166 patients with LASCCA were identified. Following ICT, 157 patients (95%) received primary curative treatment with either radiotherapy (70%), chemoradiotherapy (27%), or abdominal perineal resection (3%). The overall local tumor response rate after ICT was 76% with 20 (13%) achieving complete local tumor response. After the primary treatment, 123 patients (79%) obtained complete response, and 27 underwent salvage surgery due to persistent disease. The median follow-up time was 6 years, local and distant failure rates 22% and 13%, respectively. The 3- and 5-year disease-free survival rates were 70% and 67%, and the 3- and 5-year overall survival rates were 76% and 70%, respectively. Intensified ICT regimen could be a supplementary treatment option in the most advanced cases of LASCCA. Prospective randomized trials are needed to investigate this approach further.
Collapse
|
132
|
Lukovic J, Kim JJ, Krzyzanowska M, Chadi SA, Taniguchi CM, Hosni A. Anal Adenocarcinoma: A Rare Malignancy in Need of Multidisciplinary Management. JCO Oncol Pract 2021; 16:635-640. [PMID: 33049179 DOI: 10.1200/op.20.00363] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adenocarcinoma of the anal canal is an uncommon malignancy, making it challenging to perform randomized controlled clinical trials to define best practices in care. For patients with localized disease, there remains a lack of consensus regarding the optimal management, with some physicians advocating for trimodality therapy (similar to the locally advanced rectal adenocarcinoma paradigm) and others advocating for definitive radiation therapy with concurrent chemotherapy (similar to the management of anal squamous cell carcinoma). The objective of this clinical review is to describe the management and outcomes of patients with anal adenocarcinoma to help inform treatment recommendations.
Collapse
Affiliation(s)
- Jelena Lukovic
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - John J Kim
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Monika Krzyzanowska
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sami A Chadi
- Department of Surgical Oncology, University Health Network, Toronto, Ontario, Canada
| | - Cullen M Taniguchi
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ali Hosni
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
133
|
|
134
|
de Guillebon E, Jimenez M, Mazzarella L, Betsou F, Stadler P, Peták I, Jeannot E, Chanas L, Servant N, Marret G, Duso BA, Legrand F, Kornerup KN, Bernhart SH, Balogh G, Dóczi R, Filotás P, Curigliano G, Bièche I, Guérin J, Dirner A, Neuzillet C, Girard N, Borcoman E, Larbi Chérif L, Tresca P, Roufai DB, Dupain C, Scholl S, André F, Fernandez X, Filleron T, Kamal M, Le Tourneau C. Combining immunotherapy with an epidrug in squamous cell carcinomas of different locations: rationale and design of the PEVO basket trial. ESMO Open 2021; 6:100106. [PMID: 33865192 PMCID: PMC8066350 DOI: 10.1016/j.esmoop.2021.100106] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/12/2021] [Accepted: 03/06/2021] [Indexed: 12/25/2022] Open
Abstract
Squamous cell carcinomas (SCCs) are among the most frequent solid tumors in humans. SCCs, related or not to the human papillomavirus, share common molecular features. Immunotherapies, and specifically immune checkpoint inhibitors, have been shown to improve overall survival in multiple cancer types, including SCCs. However, only a minority of patients experience a durable response with immunotherapy. Epigenetic modulation plays a major role in escaping tumor immunosurveillance and confers resistance to immune checkpoint inhibitors. Preclinical evidence suggests that modulating the epigenome might improve the efficacy of immunotherapy. We herein review the preclinical and the clinical rationale for combining immunotherapy with an epidrug, and detail the design of PEVOsq, a basket clinical trial combining pembrolizumab with vorinostat, a histone deacetylase inhibitor, in patients with SCCs of different locations. Sequential blood and tumor sampling will be collected in order to identify predictive and pharmacodynamics biomarkers of efficacy of the combination. We also present how clinical and biological data will be managed with the aim to enable the development of a prospective integrative platform to allow secure and controlled access to the project data as well as further exploitations.
Collapse
Affiliation(s)
- E de Guillebon
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France; Inserm U932 Research Unit - Immunite et cancer, Paris, France
| | | | - L Mazzarella
- Department of Experimental Oncology, European Institute of Oncology - IRCCS, Milan, Italy; Division of Innovative Therapies, European Institute of Oncology - IRCCS, Milan, Italy
| | - F Betsou
- Integrated Biobank of Luxembourg, Dudelange, Luxembourg
| | - P Stadler
- Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany
| | - I Peták
- Oncompass Medicine Ltd, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, USA
| | - E Jeannot
- Department of Genetics, Institut Curie, Paris, France; Department of Pathology, Institut Curie, Paris, France
| | - L Chanas
- Data Direction, Institut Curie, Paris, France
| | - N Servant
- Inserm U900 Research Unit, Saint Cloud, France
| | - G Marret
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - B A Duso
- Department of Experimental Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | | | - K N Kornerup
- Integrated Biobank of Luxembourg, Dudelange, Luxembourg
| | - S H Bernhart
- Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany
| | - G Balogh
- Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany
| | - R Dóczi
- Oncompass Medicine Ltd, Budapest, Hungary
| | - P Filotás
- Oncompass Medicine Ltd, Budapest, Hungary
| | - G Curigliano
- Division of Innovative Therapies, European Institute of Oncology - IRCCS, Milan, Italy; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; University of Milano, Milan, Italy
| | - I Bièche
- Department of Genetics, Institut Curie, Paris, France
| | - J Guérin
- Data Direction, Institut Curie, Paris, France
| | - A Dirner
- Oncompass Medicine Ltd, Budapest, Hungary
| | - C Neuzillet
- Department of Medical Oncology, Institut Curie, Paris, France; Paris-Saclay University, Paris, France
| | - N Girard
- Department of Medical Oncology, Institut Curie, Paris, France; Paris-Saclay University, Paris, France
| | - E Borcoman
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - L Larbi Chérif
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - P Tresca
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - D B Roufai
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - C Dupain
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - S Scholl
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - F André
- Department of Medical Oncology, Gustave Roussy, Villejuif; INSERM, Gustave Roussy Cancer Campus, UMR981, Villejuif; University of Paris-Sud, Orsay, France
| | - X Fernandez
- Data Direction, Institut Curie, Paris, France
| | - T Filleron
- Biostatistics Unit, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - M Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France.
| | - C Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France; Inserm U900 Research Unit, Saint Cloud, France; Paris-Saclay University, Paris, France.
| |
Collapse
|
135
|
Marcus K, Lee DJ, Wilson JS, Smith RJH, Puricelli M. Pembrolizumab Induced Acute Persistent Airway Disease in a Patient with Recurrent Respiratory Papillomatosis (RRP). Ann Otol Rhinol Laryngol 2021; 131:331-336. [PMID: 34056952 DOI: 10.1177/00034894211021276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To present an uncommon but serious, recently identified complication of checkpoint inhibitor therapy in a patient treated with pembrolizumab infusion for disseminated recurrent respiratory papillomatosis (RRP). METHODS Case report. RESULTS A 43-year-old woman with underlying asthma developed acute hypoxic respiratory failure within 24 hours of her third infusion of pembrolizumab for treatment of intractable, disseminated recurrent respiratory papillomatosis. Pulmonary function testing revealed a severe intra-thoracic obstructive ventilatory defect. Discontinuation of pembrolizumab, ventilatory support, and treatment with systemic and inhaled corticosteroids resulted in resolution of respiratory failure; however, her underlying asthma remains poorly controlled. CONCLUSION To our knowledge, this case is the first report of pembrolizumab-induced obstructive respiratory failure in a patient being treated for RRP.
Collapse
Affiliation(s)
- Kathryn Marcus
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Daniel J Lee
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Jeffrey S Wilson
- Department of Internal Medicine-Pulmonology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Richard J H Smith
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Michael Puricelli
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.,Division of Otolaryngology - Head and Neck Surgery, University of Wisconsin-Madision, Madison, WI, USA
| |
Collapse
|
136
|
Wilson KC, Flood MP, Oh D, Calvin N, Michael M, Ramsay RG, Heriot AG. Immune Checkpoint Blockade in Lower Gastrointestinal Cancers: A Systematic Review. Ann Surg Oncol 2021; 28:7463-7473. [PMID: 34047860 DOI: 10.1245/s10434-021-10192-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 05/02/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Limited therapy options exist for patients with treatment-refractory metastatic colorectal or anal cancers, prompting investigation into alternative therapies. Immunotherapy in the form of immune checkpoint blockade is one such emerging treatment that has demonstrated promising results in other tumour streams.x This review aims to assess the current use of immune checkpoint blockade in patients with lower gastrointestinal tumours. PATIENTS AND METHODS Embase, Medline and Cochrane databases were searched for included studies. Clinical trials published in English and utilising immune checkpoint blockade for primary tumours situated in the lower gastrointestinal tract were included. Databases were searched for studies reporting on at least one of overall survival, progression-free survival or response to therapy. RESULTS In total, 972 abstracts were screened, with 10 studies included in the final review. Eight trials (833 patients) assessed immune checkpoint blockade in the setting of colorectal cancers. These included pembrolizumab, nivolumab, durvalumab, atezolizumab, tremelimumab and ipilimumab. A total of 20 patients across all studies achieved a complete response, and 111 patients achieved a partial response to treatment. Two trials (62 patients) assessed immune checkpoint blockade in anal cancer, utilising nivolumab and pembrolizumab. Two patients across both studies achieved a complete response, and 11 patients achieved a partial response. CONCLUSIONS A number of patients with advanced lower gastrointestinal tumours achieved a complete response to treatment for what would otherwise be considered palliative disease. Presented data have highlighted that particular patients may benefit from first-line or combination immunotherapy, and thus, further investigation is warranted to individualise treatment.
Collapse
Affiliation(s)
- K C Wilson
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Differentiation and Transcription Laboratory, Sir Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| | - M P Flood
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Differentiation and Transcription Laboratory, Sir Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - D Oh
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - N Calvin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - M Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - R G Ramsay
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Differentiation and Transcription Laboratory, Sir Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - A G Heriot
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
137
|
Eastern Canadian Gastrointestinal Cancer Consensus Conference 2019. ACTA ACUST UNITED AC 2021; 28:1988-2006. [PMID: 34073199 PMCID: PMC8161825 DOI: 10.3390/curroncol28030185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 01/06/2023]
Abstract
The annual Eastern Canadian Gastrointestinal Cancer Consensus Conference 2019 was held in Morell, Prince Edward Island, 19-21 September 2019. Experts in medical oncology, radiation oncology, and surgical oncology who are involved in the management of patients with gastrointestinal malignancies participated in presentations and discussion sessions for the purpose of developing the recommendations presented here. This consensus statement addresses multiple topics in the management of anal, colorectal, biliary tract, and gastric cancers, including: radiotherapy and systemic therapy for localized and advanced anal cancer; watch and wait strategy for the management of rectal cancer; role of testing for dihydropyrimidine dehydrogenase (DPD) deficiency prior to commencement of fluoropyrimidine therapy; radiotherapy and systemic therapy in the adjuvant and unresectable settings for biliary tract cancer; and radiotherapy and systemic therapy in the perioperative setting for early-stage gastric cancer.
Collapse
|
138
|
Hester R, Advani S, Rashid A, Holliday E, Messick C, Das P, You YQN, Taniguchi C, Koay EJ, Bednarski B, Rodriguez-Bigas M, Skibber J, Wolff R, Chang GJ, Minsky BD, Foo WC, Rothschild N, Morris VK, Eng C. CEA as a blood-based biomarker in anal cancer. Oncotarget 2021; 12:1037-1045. [PMID: 34084278 PMCID: PMC8169063 DOI: 10.18632/oncotarget.27959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The clinical utility of a blood-based biomarker in squamous cell carcinoma of the anus (SCCA) is unknown. We analyzed carcinoembryonic antigen (CEA), a commonly employed assay for patients with colorectal adenocarcinoma, as a serum biomarker for patients with biopsy-proven SCCA. MATERIALS AND METHODS Medical records from 219 patients with biopsy-proven SCCA at the University of Texas MD Anderson Cancer Center were reviewed under an IRB-approved protocol from 2013 to 2020 to assess for correlations between CEA levels and corresponding clinical and pathologic characteristics. RESULTS The mean CEA among subgroups by clinical status at the time of presentation to our institution was highest among those patients with metastatic SCCA to visceral organs (M-V, 20.7 ng/mL), however this finding was not statistically significant by ANOVA (p = .74). By clinical subgroup, the percentage of patients with an abnormally elevated CEA was highest in those patients with metastatic disease to lymph nodes (M-L, 41.2%) followed by recurrent/unresectable SCCA (36.8%), and metastatic SCCA to visceral organs (M-V, 35.2%), and was statistically significant between groups (Fisher's exact test p = .02). Using RECIST criteria for tumor progression and disease response, the mean change in CEA for patients with progression was an increase in 19 ng/mL, compared to a change of -7.3 ng/mL in those with disease response (p = .004). We likewise assessed whether CEA levels were associated with survival outcomes for all patients with metastatic SCCA, and found no correlation between CEA and likelihood for survival in a ROC analysis (multivariate, age-adjusted analysis for CEA cutoff of 8, HR = 1.01, 95% CI 0.52-1.96). CONCLUSIONS Despite interesting patterns of abnormally high CEA in SCCA patients with advanced disease, and correlation of increased CEA with disease progression (and conversely decreased CEA with disease response), CEA is not associated with survival outcomes in SCCA, and is not a clinically relevant biomarker in this disease.
Collapse
Affiliation(s)
- Robert Hester
- Division of Cancer Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
- These authors contributed equally to this work
| | - Shailesh Advani
- Division of Oncology, Terasaki Foundation of Biomedical Sciences, Los Angeles, CA, USA
- These authors contributed equally to this work
| | - Asif Rashid
- Department of Pathology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Emma Holliday
- Department of Radiation Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Craig Messick
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Yi-Qian N. You
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Cullen Taniguchi
- Department of Radiation Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Brian Bednarski
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Miguel Rodriguez-Bigas
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - John Skibber
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - George J. Chang
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Bruce D. Minsky
- Department of Radiation Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Wai Chin Foo
- Department of Pathology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole Rothschild
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Van K. Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
- These authors contributed equally to this work
| | - Cathy Eng
- Department of Hematology/Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN, USA
- These authors contributed equally to this work
| |
Collapse
|
139
|
Mattos BRS, Camandaroba MPG, Ruiz-Garcia E, Diaz-Romero C, Luca R, Mendez G, Lustosa IKF, Silva SF, Mello CA, Silva VS, O'Connor JM, Riechelmann RP. Outcomes of Patients With Metastatic Anal Cancer According to HIV Infection: A Multicenter Study by the Latin American Gastrointestinal Oncology Group (SLAGO). Clin Colorectal Cancer 2021; 20:299-304. [PMID: 34158252 DOI: 10.1016/j.clcc.2021.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND HIV-positive patients are underrepresented in clinical trials of metastatic squamous cell carcinoma of the anal canal (mSCCA). We aimed to compare the clinical outcomes of mSCCA patients according to HIV infection. METHODS This was a retrospective multicenter cohort study of consecutive patients with mSCCA. All HIV-positive patients received antiretroviral therapy. The primary endpoint was overall survival (OS), and secondary endpoints were progression-free survival (PFS) and response rate (RR). RESULTS From January 2005 to December 2019, 113 patients were included: 20 (17.6%) had HIV infection. HIV-positive patients were younger at diagnosis and more frequently male, and 20% (n = 8) received exclusively best supportive care in comparison with 8.6% of HIV-negative patients (P = .13). Both groups were similar in terms of Eastern Cooperative Oncology Group (ECOG) performance status, pattern of metastatic disease, and type of first-line chemotherapy. Five (25%) HIV-positive and 36 (38.7%) HIV-negative patients received second-line therapies (P = .24). RR and median PFS in first-line were similar between the groups: 35% and 30.1% (P = .78) and 4.9 and 5.3 months (P = .85) for patients with and without HIV infection, respectively. At a median follow-up of 26 months, median OS was 11.3 months (95% confidence interval [CI] 10.1 to 26.4) for HIV-infected patients versus 14.6 months (95% CI 11.1 to 18.1) for HIV-negative patients (P = .92). In the univariate analysis for OS, only ECOG performance status was significant. CONCLUSION HIV-positive mSCCA patients under antiretroviral therapy have oncological outcomes similar to those of HIV-negative patients. These patients should be included in trials of mSCCA.
Collapse
Affiliation(s)
- Bruna R S Mattos
- A.C. Camargo Cancer Center, Fundação Antonio Prudente, Sao Paulo, Brazil
| | | | | | | | - Romina Luca
- Institute Alexander Fleming, Buenos Aires, Argentina
| | - Guillermo Mendez
- Favaloro Foundation University Hospital, Buenos Aires, Argentina
| | - Iara K F Lustosa
- A.C. Camargo Cancer Center, Fundação Antonio Prudente, Sao Paulo, Brazil
| | - Sinara F Silva
- A.C. Camargo Cancer Center, Fundação Antonio Prudente, Sao Paulo, Brazil
| | - Celso A Mello
- A.C. Camargo Cancer Center, Fundação Antonio Prudente, Sao Paulo, Brazil
| | - Virgilio S Silva
- A.C. Camargo Cancer Center, Fundação Antonio Prudente, Sao Paulo, Brazil
| | | | | |
Collapse
|
140
|
Tostes FT, Fernandes I, Segatelli V, Callegaro D, Carmagnani Pestana R. Response to Pembrolizumab in Advanced Anal Squamous Cell Carcinoma With High TMB and PD-L1 and PD-L2 Amplification. Clin Colorectal Cancer 2021; 20:350-353. [PMID: 34154923 DOI: 10.1016/j.clcc.2021.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 02/08/2023]
Abstract
We report the case of a 44-year-old female with a prior diagnosis of Sjögren's syndrome who was treated for metastatic anal squamous cell carcinoma with second-line pembrolizumab and has achieved a sustained partial response after a follow-up of 13 months. Comprehensive genomic profiling was remarkable for PD-L1 and PD-L2 amplification and a high tumor mutational burden (19 mutations per megabase). To the best of our knowledge, we present the first report to correlate PD-L1 and PD-L2 amplification with good outcomes of immune checkpoint inhibition in metastatic anal squamous cell carcinoma.
Collapse
Affiliation(s)
| | - Italo Fernandes
- Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval
| | - Vanderlei Segatelli
- Departamento de Patologia, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Donato Callegaro
- Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval
| | | |
Collapse
|
141
|
Wakeham K, Murray L, Muirhead R, Hawkins MA, Sebag-Montefiore D, Brown S, Murphy L, Thomas G, Bell S, Whibley M, Morgan C, Sleigh K, Gilbert DC. Multicentre Investigation of Prognostic Factors Incorporating p16 and Tumour Infiltrating Lymphocytes for Anal Cancer After Chemoradiotherapy. Clin Oncol (R Coll Radiol) 2021; 33:638-649. [PMID: 34024700 DOI: 10.1016/j.clon.2021.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022]
Abstract
AIMS Anal squamous cell carcinomas (ASCC) are strongly associated with human papillomaviruses. Standard of care is chemoradiotherapy at uniform doses with no treatment stratification. Immunohistochemical staining for p16INK4A (p16), a surrogate for human papillomaviruses, is prognostic for outcomes. We investigated this alongside clinical-pathological factors, including tumour infiltrating lymphocyte (TIL) scores. MATERIALS AND METHODS Using an independent, multicentre cohort of 257 ASCC treated with chemoradiotherapy, pretreatment biopsies were stained and scored for p16 and TIL. Kaplan-Meier curves were derived for outcomes (disease-free survival [DFS], overall survival and cancer-specific survival), by stage, p16 and TIL scores and Log-rank tests were carried out to investigate prognostic effect. A multivariate analysis was carried out using Cox regression. RESULTS Stage, sex, p16 and TILs were independently prognostic. Hazard ratios for death (overall survival) were 2.51 (95% confidence interval 1.36-4.63) for p16 negative versus p16 positive, 2.17 (1.34-3.5) for T3/4 versus T1/2, 2.42 (1.52-3.8) for males versus females and 3.30 (1.52-7.14) for TIL1 versus TIL3 (all P < 0.05). CONCLUSIONS We have refined prognostic factors in ASCC. p16 adds to stratification by stage with respect to DFS in early disease and overall survival/DFS in locally advanced cancers. Our data support the role of the host immune response in mediating outcomes. These factors will be prospectively evaluated in PLATO (ISRCTN88455282).
Collapse
Affiliation(s)
- K Wakeham
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - L Murray
- Leeds Institute of Medical Research, University of Leeds, Leeds Cancer Centre, Leeds, UK
| | - R Muirhead
- Oxford University Hospitals NHS Trust, Department of Oncology, Churchill Hospital, Oxford, UK
| | - M A Hawkins
- University College London, Medical Physics and Biomedical Engineering, London, UK
| | - D Sebag-Montefiore
- Leeds Institute of Medical Research, University of Leeds, Leeds Cancer Centre, Leeds, UK
| | - S Brown
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | - L Murphy
- MRC Clinical Trials Unit at UCL, London, UK
| | - G Thomas
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - S Bell
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - M Whibley
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, UK
| | - C Morgan
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, UK
| | - K Sleigh
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, UK
| | - D C Gilbert
- MRC Clinical Trials Unit at UCL, London, UK.
| |
Collapse
|
142
|
Ouyang T, Cao Y, Kan X, Chen L, Ren Y, Sun T, Yan L, Xiong B, Liang B, Zheng C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front Oncol 2021; 11:621639. [PMID: 34046338 PMCID: PMC8144509 DOI: 10.3389/fonc.2021.621639] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immune Checkpoint Inhibitors (ICI) have been progressively used in cancer treatment and produced unique toxicity profiles. This systematic review aims to comprehend the patterns and occurrence of treatment-related adverse events (trAEs) based on ICI. Methods PICOS/PRISMA methods were used to identify published English-language on PubMed, Web of Science, and Scopus from 2015 to 2020. Published clinical trials on ICI monotherapy, combined ICIs, and ICI plus other treatment with tabulated data on grade≥3 trAEs were included. Odds ratio (OR), χ2 tests were used to analyze for effect size and associations. Results This review included 145 clinical trials involving 21786 patients. Grade 3-5 trAEs were more common with ICI when they were plused with other treatments compared with ICI monotherapy(54.3% versus 17.7%, 46.1%, p<0.05). Grade 3-5 trAEs were also more common with CTLA-4 mAbs compared with anti-PD-1 and anti-PD-L1 (34.2% versus 15.1%, 13.6%, p<0.05). Hyperthyroidism (OR 3.8, 95%CI 1.7–8.6), nausea (OR 3.7, 95%CI 2.5–5.3), diarrhea (OR 2.7, 95%CI 2.2–3.2), colitis (OR 3.4, 95%CI 2.7–4.3), ALT increase (OR 4.9, 95%CI 3.9–6.1), AST increase (OR 3.8, 95%CI 3.0–4.9), pruritus (OR 2.4, 95%CI 1.5–3.9), rash (OR 2.8, 95%CI 2.1–3.8), fatigue (OR 2.8, 95%CI 2.2–3.7), decreased appetite (OR 2.4, 95%CI 1.5–3.8), and hypophysitis (OR 2.0, 95%CI 1.2–3.3) were more frequent with combined ICIs. Diarrhea (OR 8.1, 95%CI 6.4–10.3), colitis (OR 12.2, 95%CI 8.7–17.1), ALT increase (OR 5.1, 95%CI 3.5–7.4), AST increase (OR 4.2, 95%CI 2.8–6.3), pruritus (OR 4.1, 95%CI 2.0–8.4), rash (OR 4.4, 95%CI 2.9–6.8), hypophysitis (OR 12.1, 95%CI 6.3–23.4) were more common with CTLA-4 mAbs; whereas pneumonitis (OR 4.7, 95% CI 2.1–10.3) were more frequent with PD-1 mAbs. Conclusions Different immune checkpoint inhibitors are associated with different treatment-related adverse events profiles. A comprehensive data in this systematic review will provide comprehensive information for clinicians.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Liangliang Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
143
|
Livingston AJ, Bailey CE, Washington MK, Eng C. Squamous Cell Carcinoma of the Anal Verge with Sigmoid Colon Metastasis. Clin Colorectal Cancer 2021; 20:e210-e213. [PMID: 34092493 DOI: 10.1016/j.clcc.2021.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Austin J Livingston
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN.
| | - Christina E Bailey
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Cathy Eng
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
144
|
de Almeida NAA, Ribeiro CRDA, Raposo JV, de Paula VS. Immunotherapy and Gene Therapy for Oncoviruses Infections: A Review. Viruses 2021; 13:822. [PMID: 34063186 PMCID: PMC8147456 DOI: 10.3390/v13050822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has been shown to be highly effective in some types of cancer caused by viruses. Gene therapy involves insertion or modification of a therapeutic gene, to correct for inappropriate gene products that cause/may cause diseases. Both these types of therapy have been used as alternative ways to avoid cancers caused by oncoviruses. In this review, we summarize recent studies on immunotherapy and gene therapy including the topics of oncolytic immunotherapy, immune checkpoint inhibitors, gene replacement, antisense oligonucleotides, RNA interference, clustered regularly interspaced short palindromic repeats Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based gene editing, transcription activator-like effector nucleases (TALENs) and custom treatment for Epstein-Barr virus, human T-lymphotropic virus 1, hepatitis B virus, human papillomavirus, hepatitis C virus, herpesvirus associated with Kaposi's sarcoma, Merkel cell polyomavirus, and cytomegalovirus.
Collapse
Affiliation(s)
| | | | | | - Vanessa Salete de Paula
- Laboratory of Molecular Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, 21040-360 Rio de Janeiro, Brazil; (N.A.A.d.A.); (C.R.d.A.R.); (J.V.R.)
| |
Collapse
|
145
|
Molecular and immunophenotypic characterization of anal squamous cell carcinoma reveals distinct clinicopathologic groups associated with HPV and TP53 mutation status. Mod Pathol 2021; 34:1017-1030. [PMID: 33483624 DOI: 10.1038/s41379-020-00729-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/30/2022]
Abstract
Squamous cell carcinoma (SqCC) is the most common malignancy of the anal canal, where it is strongly associated with HPV infection. Characteristic genomic alterations have been identified in anal SqCC, but their clinical significance and correlation with HPV status, pathologic features, and immunohistochemical markers are not well established. We examined the molecular and clinicopathologic features of 96 HPV-positive and 20 HPV-negative anal SqCC. HPV types included 89 with HPV16, 2 combined HPV16/HPV18, and 5 HPV33. HPV-positive cases demonstrated frequent mutations or amplifications in PIK3CA (30%; p = 0.027) or FBXW7 mutations (10%). HPV-negativity was associated with frequent TP53 (53%; p = 0.00001) and CDKN2A (21%; p = 0.0045) mutations. P16 immunohistochemistry was positive in all HPV-positive cases and 3/20 HPV-negative cases (p < 0.0001; sensitivity: 100%; specificity: 85%) and was associated with basaloid morphology (p = 0.0031). Aberrant p53 immunohistochemical staining was 100% sensitive and specific for TP53 mutation (p < 0.0001). By the Kaplan-Meier method, HPV-negativity, aberrant p53 staining, and TP53 mutation were associated with inferior overall survival (OS) (p < 0.0001, p = 0.0103, p = 0.0103, respectively) and inferior recurrence-free survival (p = 0.133, p = 0.0064, and p = 0.0064, respectively). TP53/p53 status stratified survival probability by HPV status (p = 0.013), with HPV-negative/aberrant p53 staining associated with the worst OS, HPV-positive/wild-type p53 with best OS, and HPV-positive/aberrant p53 or HPV-negative/wild-type p53 with intermediate OS. On multivariate analysis HPV status (p = 0.0063), patient age (p = 0.0054), T stage (p = 0.039), and lymph node involvement (p = 0.044) were independently associated with OS. PD-L1 expression (CPS ≥ 1) was seen in 30% of HPV-positive and 40% of HPV-negative cases, and PD-L1 positivity was associated with a trend toward inferior OS within the HPV-negative group (p = 0.064). Our findings suggest that anal SqCC can be subclassified into clinically, pathologically, and molecularly distinct groups based on HPV and TP53 mutation status, and p16 and p53 immunohistochemistry represent a clinically useful method of predicting these prognostic groups.
Collapse
|
146
|
Carr RM, Jin Z, Hubbard J. Research on Anal Squamous Cell Carcinoma: Systemic Therapy Strategies for Anal Cancer. Cancers (Basel) 2021; 13:cancers13092180. [PMID: 34062753 PMCID: PMC8125190 DOI: 10.3390/cancers13092180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Anal cancer is rare with an estimated 9000 new cases predicted to occur in the United States in 2021. However, rates of new anal cancer cases and deaths from the disease are increasing by about 2% and 3% per year respectively. In light of these trends it is critical to better understand the nature of this disease and progress in its management. The present review focuses on the history and development of the role of systemic therapy in the treatment of anal cancer. Major trials establishing the role of chemotherapy in the management of locoregional and metastatic anal cancer are summarized. In addition, the rapidly evolving role of immunotherapy is discussed. Finally, major insights into the molecular pathobiology of anal cancer and opportunities for advancement in precision medicine in treatment of the disease. Abstract Anal squamous cell carcinoma (ASCC) is a rare malignancy, with most cases associated with human papilloma virus and an increased incidence in immunocompromised patients. Progress in management of ASCC has been limited not only due to its rarity, but also the associated lack of research funding and social stigma. Historically, standard of care for invasive ASCC has been highly morbid surgical resection, requiring a permanent colostomy. Surgery was associated with disease recurrence in approximately half of the patients. However, the use of chemotherapy (5-fluorouracil and mitomycin C) concomitantly with radiation in the 1970s resulted in disease regression, curing a subset of patients and sparing them from morbid surgery. Validation of the use of systemic therapy in prospective trials was not achieved until approximately 20 years later. In this review, advancements and shortcomings in the use of systemic therapy in the management of ASCC will be discussed. Not only will standard-of-care systemic therapies for locoregional and metastatic disease be reviewed, but the evolving role of novel treatment strategies such as immune checkpoint inhibitors, HPV-based vaccines, and molecularly targeted therapies will also be covered. While advances in ASCC treatment have remained largely incremental, with increased biological insight, an increasing number of promising systemic treatment modalities are being explored.
Collapse
|
147
|
Cimino SK, Ciombor KK, Chakravarthy AB, Bailey CE, Hopkins MB, Geiger TM, Hawkins AT, Eng C. Safety considerations with new treatment regimens for anal cancer. Expert Opin Drug Saf 2021; 20:889-902. [PMID: 33900857 DOI: 10.1080/14740338.2021.1915281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Anal cancer is a rare malignancy, but incidence rates are rising. Primary chemoradiation is the standard of care for early disease with surgery reserved for salvage. Despite success in terms of survival, patients suffer significant morbidity. Research is underway to advance the field and improve outcomes for these patients.Areas covered: This review aims to discuss the safety and efficacy of new approaches to treat anal cancer. A literature search was performed from January 1950 through November 2020 via PubMed and ClinicalTrials.gov databases to obtain data from ongoing or published studies examining new regimens for the treatment of anal cancers. Pertinent topics covered include miniature drug conjugates, epidermal growth factor receptor inhibitors, checkpoint inhibitor combinations, and novel immunomodulators.Expert opinion: Based on emerging clinical data, the treatment paradigm for anal cancer is likely to shift in the upcoming years. One of the largest areas of investigation is the field of immunotherapy, which may emerge as an integral component of anal cancer for all treatment settings.
Collapse
Affiliation(s)
- Sarah K Cimino
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kristen K Ciombor
- Department of Medicine: Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A Bapsi Chakravarthy
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christina E Bailey
- Department of Surgery: Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Benjamin Hopkins
- Department of Surgery: Division of Colon and Rectal Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy M Geiger
- Department of Surgery: Division of Colon and Rectal Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander T Hawkins
- Department of Surgery: Division of Colon and Rectal Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cathy Eng
- Department of Medicine: Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
148
|
Fu J, Li WZ, McGrath NA, Lai CW, Brar G, Xiang YQ, Xie C. Immune Checkpoint Inhibitor Associated Hepatotoxicity in Primary Liver Cancer Versus Other Cancers: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:650292. [PMID: 33968750 PMCID: PMC8097087 DOI: 10.3389/fonc.2021.650292] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Overall risks of hepatotoxicity with immune checkpoint inhibitors (ICIs) have yet to be compared in primary liver cancers to other solid tumors. METHODS We reviewed data from the PubMed, Embase, and Scopus databases, and assessed the risk of hepatotoxicity associated with ICIs. RESULTS A total of 117 trials were eligible for the meta-analysis, including 7 trials with primary liver cancers. The most common hepatotoxicity was ALT elevation (incidence of all grade 5.29%, 95% CI 4.52-6.20) and AST elevation (incidence of all grade 5.88%, 95% CI 4.96-6.97). The incidence of all grade ALT and AST elevation was 6.01% and 6.84% for anti-PD-1 (95% CI 5.04-7.18/5.69-8.25) and 3.60% and 3.72% for anti-PD-L1 (95% CI 2.72-4.76/2.82-4.94; p< 0.001/p<0.001). The incidence of ≥ grade 3 ALT and AST elevation was 1.54% and 1.48% for anti-PD-1 (95% CI 1.19-1.58/1.07-2.04) and 1.03% and 1.08% for anti-PD-L1 (95% CI 0.71-1.51/0.80-1.45; p= 0.002/p<0.001). The incidence of all grade ALT and AST elevation was 13.3% and 14.2% in primary liver cancers (95% CI 11.1-16.0 and 9.93-20.36) vs. 4.92% and 5.38% in other solid tumors (95% CI 4.21-5.76 and 4.52-5.76 in other solid tumors; p <0.001/p<0.001). CONCLUSION Our study indicates that anti-PD-1 is associated with a higher risk of all- and high-grade hepatotoxicity compared to anti-PD-L1, and primary liver cancers are associated with a higher risk of all- and high-grade hepatotoxicity compared to other solid tumors.
Collapse
Affiliation(s)
- Jianyang Fu
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wang-Zhong Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Nicole A. McGrath
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Chunwei Walter Lai
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Gagandeep Brar
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Changqing Xie
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
149
|
Patel SP, Mayerson E, Chae YK, Strosberg J, Wang J, Konda B, Hayward J, McLeod CM, Chen HX, Sharon E, Othus M, Ryan CW, Plets M, Blanke CD, Kurzrock R. A phase II basket trial of Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors (DART) SWOG S1609: High-grade neuroendocrine neoplasm cohort. Cancer 2021; 127:3194-3201. [PMID: 33882143 DOI: 10.1002/cncr.33591] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND The authors previously reported the results of the nonpancreatic neuroendocrine neoplasm cohort of the SWOG S1609 DART (Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors) trial, which permitted all histologic grades and had a 44% overall response rate (ORR) among patients with high-grade disease. Here they sought to validate their findings in a dedicated prospective cohort of high-grade neuroendocrine neoplasms within S1609. METHODS A prospective, open-label, multicenter, phase 2 clinical trial of ipilimumab plus nivolumab was conducted across multiple rare tumor cohorts. The dedicated, high-grade neuroendocrine neoplasm cohort was examined here. The primary end point was the ORR according to version 1.1 of the Response Evaluation Criteria in Solid Tumors. Secondary end points included progression-free survival (PFS), overall survival (OS), and toxicity. RESULTS Nineteen patients with high-grade neuroendocrine neoplasms (defined by local pathology review) were enrolled in this cohort of S1609. The most common primary sites were unknown primaries (21%), which were followed by the rectum, gastroesophageal junction, cervix, and pancreas (11%). The median number of lines of prior therapy was 1 (range, 0-3). All patients were microsatellite-stable. The median Ki-67 value was 80%. The ORR was 26% (95% confidence interval [CI], 11%-45%), and the clinical benefit rate (stable disease for ≥6 months plus partial responses plus complete responses) was 32% (95% CI, 13%-57%). The 6-month PFS rate was 32% (95% CI, 16%-61%) with a median PFS of 2.0 months (95% CI, 1.8 months to ∞) and a median OS of 8.7 months (95% CI, 6.1 months to ∞). The most common toxicities were fatigue (32%) and rash (26%), and the most common grade 3/4 immune-related adverse event was rash (15%); there were no events that required treatment discontinuation and no grade 5 events. CONCLUSIONS Ipilimumab plus nivolumab demonstrated a 26% ORR in patients with high-grade neuroendocrine neoplasms, with durable responses seen in patients with refractory disease.
Collapse
Affiliation(s)
- Sandip Pravin Patel
- Moores Cancer Center, University of California at San Diego, La Jolla, California
| | - Edward Mayerson
- SWOG Statistical Center, Seattle, Washington.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | - Jue Wang
- University of Arizona, Phoenix, Arizona
| | - Bhavana Konda
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jourdain Hayward
- SWOG Data Operations Center/Cancer Research and Biostatistics, Seattle, Washington
| | - Christine M McLeod
- SWOG Data Operations Center/Cancer Research and Biostatistics, Seattle, Washington
| | - Helen X Chen
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Elad Sharon
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Megan Othus
- SWOG Statistical Center, Seattle, Washington.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Melissa Plets
- SWOG Statistical Center, Seattle, Washington.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Charles D Blanke
- SWOG Group Chair's Office, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Razelle Kurzrock
- Moores Cancer Center, University of California at San Diego, La Jolla, California
| |
Collapse
|
150
|
Chen S, Zhang Z, Zheng X, Tao H, Zhang S, Ma J, Liu Z, Wang J, Qian Y, Cui P, Huang D, Huang Z, Wu Z, Hu Y. Response Efficacy of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:562315. [PMID: 33937012 PMCID: PMC8085334 DOI: 10.3389/fonc.2021.562315] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Background Immune checkpoint inhibitors targeting the PD-1/PD-L1 pathway have demonstrated promise in treating a variety of advanced cancers; however, little is known regarding their efficacy under various clinical situations, including different cancer types, treatment lines, drug combinations, and therapeutic regimens. Methods Published articles and conference abstracts (in English) in PubMed, Embase, the Cochrane Central Register, and Web of Science were searched up to February 10, 2020. The data were analyzed by the meta-analysis program in Stata. Results A total of 16,400 patients from 91 clinical trials were included in this meta-analysis. PD-1/PD-L1 inhibitors had a mean ORR of 19.56% (95% CI: 15.09–24.03), a median TTR of 2.05 months (m) (95%CI: 1.85–2.26), and a median DOR of 10.65 m (95%CI: 7.78–13.52). First-line treatment had a higher ORR (36.57% vs. 13.18%) but a shorter DOR (9.00 m vs. 13.42 m) compared to the second-line or subsequent treatment. Immunotherapy combined with chemotherapy (I+C) (46.81% [95%CI: 36.02–57.60]) had a statistically significant higher ORR compared to immunotherapy (I) (17.75% [95%CI: 14.47–21.03]) or immunotherapy combined with immunotherapy (I+O) (12.25% [95%CI: 1.56–22.94]), while I+C (8.09 m [95%CI: 6.86–9.32]) appeared to reduce the DOR compared to I (12.39 m [95%CI: 7.60–17.18]). PD-1 inhibitors were associated with better ORR (21.65% vs. 17.60%) and DOR (11.26 m vs. 10.03 m) compared to PD-L1 inhibitors. There were no significant differences in TTR under different situations. Conclusions PD-1/PD-L1 inhibitors were promising immunotherapeutic agents to achieve satisfactory response efficacies with different cancer types, treatment lines, drug combinations, and therapeutic regimens. This comprehensive summary of the response efficacy of PD-1/PD-L1 inhibitors serves as a reference for clinicians to make evidence-based decisions.
Collapse
Affiliation(s)
- Shixue Chen
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Zhibo Zhang
- Department of Cardiothoracic Surgery, The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Xuan Zheng
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Haitao Tao
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Sujie Zhang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Junxun Ma
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhefeng Liu
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jinliang Wang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuanyu Qian
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Pengfei Cui
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Di Huang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Ziwei Huang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Zhaozhen Wu
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Yi Hu
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|