1
|
Liao Y, Wang D, Yang X, Ni L, Lin B, Zhang Y, Feng G, Li J, Gao F, Liao M, Du X, Chen W. High‑intensity focused ultrasound thermal ablation boosts the efficacy of immune checkpoint inhibitors in advanced cancers with liver metastases: A single‑center retrospective cohort study. Oncol Lett 2025; 29:124. [PMID: 39807097 PMCID: PMC11726302 DOI: 10.3892/ol.2025.14871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
High-intensity focused ultrasound thermal ablation (HIFU) is a novel non-invasive technique in the treatment of liver metastases (LIM) that allows focal destruction and is not affected by dose limits. This retrospective study aimed to explore the efficacy of HIFU in improving survival and the safety of the method in newly diagnosed patients with cancer with LIM who received first-line immune checkpoint inhibitor (ICI) therapy. Between January 2018 and December 2023, data from 438 newly diagnosed patients with cancer and LIM who were treated at Mianyang Central Hospital (Mianyang, China) were reviewed. A total of 94 patients were enrolled in this study, of whom 28 were diagnosed with lung carcinoma, 36 with gastric carcinoma, 11 with esophageal carcinoma, 7 with cholangiocarcinoma and 12 with other malignancies. The patients were divided into groups depending on whether they underwent HIFU. Progression-free survival (PFS), overall survival (OS) and adverse events (AEs) were compared. Clinicopathological features were analyzed using the chi-squared test. Of the 94 patients, 28 received ICI + HIFU as first-line treatment. After a median follow-up of 13.8 months, the median PFS and OS in the HIFU group were 2.38 times [10.95 vs. 4.60 months, 95% confidence interval (CI): 1.087-3.106, P<0.0001] and 1.84 times (19.6 vs. 10.67 months, 95% CI: 1.087-3.106, P=0.0418), respectively, higher than in the group without HIFU. All-cause AEs and immune-mediated AEs were similar between the groups with and without HIFU. However, the incidence of grade 1-2 immune-mediated AEs, troponin elevation, hepatotoxicity and renal dysfunction were more common in the current patients with LIM than those reported previously for the entire population. No immune-mediated AEs of grade ≥3 occurred in either group. HIFU prolonged the PFS and OS of first-line ICI in newly diagnosed patients with advanced cancer with LIM, with manageable safety and tolerability. The efficacy of HIFU in patients with LIM who plan to undergo ICI treatment warrants further prospective clinical investigation.
Collapse
Affiliation(s)
- Yao Liao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, P.R. China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Decai Wang
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Xiyue Yang
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Lu Ni
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Binwei Lin
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Yu Zhang
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Gang Feng
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Jie Li
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Feng Gao
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
- Department of Oncology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Min Liao
- Department of Information, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Xiaobo Du
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Wenzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
2
|
Gueiderikh A, Faivre JC, Golfier C, Escande A, Thureau S. Efficacy of innovative systemic treatments in combination with radiotherapy for bone metastases: a GEMO (the European Study Group of Bone Metastases) state of the art. Cancer Metastasis Rev 2025; 44:28. [PMID: 39875680 PMCID: PMC11775081 DOI: 10.1007/s10555-024-10236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
The management of bone metastases (BoM) requires a multidisciplinary approach to prevent complications, necessitating updated knowledge in light of the rapid advancements in systemic treatments and surgical, interventional radiology or radiation techniques. This review aims to discuss efficacy of new systemic treatments on BoM, the benefits of radiotherapy adjunction, and the optimal methods for combining them. Preliminary evidence suggesting reduced efficacy of immune checkpoint inhibitors (ICI), and several multi-kinase inhibitors regarding BoM may encourage early use of radiotherapy (RT). Systemic treatment efficacy modulation by RT and ablative RT strategies are explored. Concerns for increased side effects for several kinase inhibitors and double ICI in combination with RT imply suspending those systemic treatments during RT. Various timing strategies to combine prostate hormone therapies and RT are developed. Emerging internal vectorized radiotherapy molecules necessitate developing new combination strategies with RT. Further prospective data collection and comparative trials should be encouraged.
Collapse
Affiliation(s)
- Anna Gueiderikh
- Département de Radiothérapie, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Orsay, France
| | - Jean-Christophe Faivre
- Radiation Oncology Department, Institut de Cancérologie de Lorraine, Vandoeuvre-Lès-Nancy, 54519, France
| | - Constance Golfier
- Radiation Oncology Department, Institut de Cancérologie de Lorraine, Vandoeuvre-Lès-Nancy, 54519, France
| | - Alexandre Escande
- Service de Radiothérapie, Centre Léonard de Vinci, Dechy, France
- Laboratoire CRIStAL, UMR 9186, Université de Lille, Lille, France
- Faculté de Médecine H.Warembourg, Université de Lille, Lille, France
| | - Sébastien Thureau
- Département de Radiothérapie et de Physique Médicale, Centre Henri Becquerel Rouen QuantiF, LITIS EA4108 Université Rouen, Rouen, France.
| |
Collapse
|
3
|
Li Y, Min Y, Wei Z, Liu Z, Pei Y, Yang Y, Gao K, Song G, Xu S, He S, Ge J, Qing Y, Wei Y, Ai P, Chen Y, Peng X. Metastatic sites of baseline as predictors in recurrent or metastatic nasopharyngeal carinoma treated with PD-L1 inhibitor: a secondary analysis of multicenter, single-arm, phase II study (KL-A167). Cancer Immunol Immunother 2025; 74:72. [PMID: 39751901 PMCID: PMC11699008 DOI: 10.1007/s00262-024-03905-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) show optimal treatment effects on recurrent or metastatic nasopharyngeal carcinoma(R/M NPC). Nonetheless, whether metastatic sites impact ICIs efficacy remains unclear. METHODS We performed a secondary analysis of R/M NPC patients treated with KL-A167, a programmed cell death-ligand 1(PD-L1) inhibitor, based on a multicenter, single-arm, phase II study from China between 2019 and 2021 years, which represents the first and most comprehensive analysis of the effectiveness of a PD-L1 inhibitor in patients who have been previously treated. The Cox proportional hazard model was utilized to evaluate the association between sites and PFS and OS. Sensitivity analysis and subgroup analysis were carried out to confirm the reliability of our findings. RESULTS A total of 153 R/M NPC patients were included. The mean age was 47 years and 81% of patients were males. All patients in our study had distant metastasis, with a majority (n = 69) presenting with more than 2 sites of distant metastasis upon admission. The collected sites of metastasis included liver, lung, lymph and bone. Among the 153 patients, 37.9% (58 patients) received anti-PD-L1 treatment for a minimum of 6 months, and 17.6% (27 patients) were treated for at least 12 months. By conducting multivariate analysis, R/M NPC patients with non-liver metastases presented significantly longer progress-free survival (PFS, HR:1.67, CI:1.09-0.2.55, p = 0.018) and overall survival (OS, HR:2.52, CI:1.49-4.28, p < 0.001) compared with those with liver metastasis. The median PFS (72 vs. 144 days, p < 0.0001) and OS (730 vs. 305 days, p < 0.0001) were significantly longer for patients with non-liver metastases. However, lung, bone and lymph node metastasis had no statistical significance on PFS and OS (p > 0.005). Our sensitive analysis showed liver metastases patients with less other site metastases (0 or 1) had shorter OS compared to non-liver metastases patients with more other metastases(≥ 2). Furthermore, subgroup analysis indicated the robustness evidence liver metastasis indeed a valuable prognostic factor for survival. CONCLUSIONS Compared to patients with other metastatic sites, R/M NPC patients with liver metastasis have poor survival patterns when receiving anti-PD-L1 therapy. Our study provides rational evidence for the urgent need to explore more efficacy treatment modalities for NPC patients with liver metastasis.
Collapse
Affiliation(s)
- Yuantai Li
- Division of Head & Neck Tumor Multimodality Treatment, Department of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, China
| | - Yu Min
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China
| | - Zhigong Wei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China
| | - Yiyan Pei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China
| | - Yujie Yang
- Department of Intensive Care Medicine, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kun Gao
- Division of Head & Neck Tumor Multimodality Treatment, Department of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, China
| | - Ge Song
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China
| | - Shihong Xu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China
| | - Shuangshuang He
- Division of Head & Neck Tumor Multimodality Treatment, Department of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, China
| | - Junyou Ge
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Chengdu, Sichuan, China
| | - Yan Qing
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Chengdu, Sichuan, China
| | - Youneng Wei
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Chengdu, Sichuan, China
| | - Ping Ai
- Division of Head & Neck Tumor Multimodality Treatment, Department of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, China.
| | - Ye Chen
- Department of Abdominal Cancer, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Sichuan, 610041, Chengdu, China.
| |
Collapse
|
4
|
Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025:10.1038/s41577-024-01111-8. [PMID: 39748116 DOI: 10.1038/s41577-024-01111-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Cancers can avoid immune-mediated elimination by acquiring traits that disrupt antitumour immunity. These mechanisms of immune evasion are selected and reinforced during tumour evolution under immune pressure. Some immunogenic subclones are effectively eliminated by antitumour T cell responses (a process known as immunoediting), which results in a clonally selected tumour. Other cancer cells arise to resist immunoediting, which leads to a tumour that includes several distinct cancer cell populations (referred to as intratumour heterogeneity (ITH)). Tumours with high ITH are associated with poor patient outcomes and a lack of responsiveness to immune checkpoint blockade therapy. In this Review, we discuss the different ways that cancer cells evade the immune system and how these mechanisms impact immunoediting and tumour evolution. We also describe how subclonal antigen presentation in tumours with high ITH can result in immune evasion.
Collapse
Affiliation(s)
- Malte Roerden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Ragon Institute of Mass General Hospital, Massachusetts Institute for Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Ćeriman Krstić V, Samardžić N, Gajić M, Savić M, Šeha B, Roksandić Milenković M, Jovanović D. Treatment Options for Patients with Non-Small Cell Lung Cancer and Liver Metastases. Curr Issues Mol Biol 2024; 46:13443-13455. [PMID: 39727930 PMCID: PMC11726995 DOI: 10.3390/cimb46120802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Lung cancer represents the most common cause of cancer-related death. Patients with non-small cell lung cancer (NSCLC) and liver metastases have worse prognosis, with an overall survival (OS) from three to six months. The majority of them have a poor response to chemotherapy, and the data are controversial regarding the response to immunotherapy. This could be because the liver is considered to be an immune-tolerant organ, which is characterized by T-cell anergy and immunosuppressive signals. This review evaluates current treatment options for patients with NSCLC and liver metastases. Combination therapies might be a better treatment option for this subgroup of patients. The addition of radiotherapy to immunotherapy could also be an option in selected patients. The resection of single liver metastasis should also be considered.
Collapse
Affiliation(s)
- Vesna Ćeriman Krstić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (N.S.); (M.G.)
| | - Natalija Samardžić
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (N.S.); (M.G.)
| | - Milija Gajić
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (N.S.); (M.G.)
| | - Milan Savić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Clinic for Thoracic Surgery, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Biljana Šeha
- Clinic for Neurosurgery, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | | | | |
Collapse
|
6
|
Wang L, Liu SS, Zhang SM, Chen XQ, Huang T, Tian R, Zhao YQ, Chen Z, Xianba CR. Gastric cancer liver metastasis will reduce the efficacy of immunotherapy. World J Gastrointest Surg 2024; 16:2760-2764. [PMID: 39351566 PMCID: PMC11438812 DOI: 10.4240/wjgs.v16.i9.2760] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/06/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
Immune checkpoint inhibitors augment the antitumor activity of T cells by inhibiting the negative regulatory pathway of T cells, leading to notable efficacy in patients with non-small cell lung cancer, melanoma, and other malignancies through immunotherapy utilization. However, secondary malignant liver tumors not only lower the liver's sensitivity to immunotherapy but also trigger systemic immune suppression, resulting in reduced overall effectiveness of immune therapy. Patients receiving immunotherapy for non-small cell lung cancer and melanoma experience reduced response rates, progression-free survival, and overall survival when secondary malignant tumors develop in the liver. Through Liu's retrospective analysis, valuable insights are provided for the future clinical management of these patients. Therefore, in patients with gastric cancer (GC), the occurrence of liver metastasis might be indicative of reduced efficacy of immunotherapy. Overcoming liver immune tolerance mechanisms and their negative impacts allows for the potential benefits of immunotherapy in patients with GC and liver metastasis.
Collapse
Affiliation(s)
- Liang Wang
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Shan-Shan Liu
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Sheng-Mei Zhang
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Xiao-Qian Chen
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Tao Huang
- Intensive Care Unit, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Rong Tian
- Department of Ultrasound, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Ya-Qi Zhao
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Zhou Chen
- Department of Gastrointestinal Oncology Surgery, The Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, Qinghai Province, China
| | - Cai-Rang Xianba
- Department of General Surgery, Hainan State People's Hospital of Qinghai Province, Hainan Tibetan Autonomous Prefecture 813000, Qinghai Province, China
| |
Collapse
|
7
|
Cheon J, Jung S, Kim JS, Kang B, Kim H, Chan LL, Becker L, Gaillard VE, Chan SL, Kim C, Chon HJ. Organ-specific responses to atezolizumab plus bevacizumab in advanced hepatocellular carcinoma: A multicentre, retrospective study. Liver Int 2024; 44:1961-1970. [PMID: 38618972 DOI: 10.1111/liv.15935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND AND AIMS Anti-programmed death 1 (PD-1) monotherapy triggers various responses by each organ. In advanced hepatocellular carcinoma (HCC), while extrahepatic lesions demonstrate objective response rates (ORR) of 20%-40%, only 10% of intrahepatic lesions respond. Although first-line atezolizumab/bevacizumab has shown survival benefits in advanced HCC, organ-specific responses remain unexplored. Therefore, we aimed to assess organ-specific responses in patients with advanced HCC receiving atezolizumab/bevacizumab. METHODS This retrospective, multicenter, observational study included patients who received first-line atezolizumab/bevacizumab for advanced HCC. Patients with Child-Pugh class A, measurable tumour lesions and serial imaging available for response evaluation were eligible. RESULTS Between May 2020 and June 2021, 131 patients (median age: 62) from three cancer referral institutions were included. Ninety-one had hepatitis B (69.5%), 108 were at Barcelona clinic liver cancer stage C (82.4%), and 78 had extrahepatic metastasis (59.5%). After a median follow-up of 10.1 months, median progression-free survival was 6.8 months (95% confidence interval [CI], 4.6-9.2), median overall survival remained unreached (95% CI, range unavailable) and the ORR was 29.0%. Among 270 individual tumour lesions, the liver was the most commonly involved organ (n = 158). Atezolizumab/bevacizumab induced ORR of 27.8%, 42.2%, 29.1% and 21.0% for liver, lymph nodes, lungs and other sites, respectively. The organ-specific response rate for intrahepatic tumours decreased with increasing size (35.6%: <5 cm, 15.0%: ≥ 5 cm). CONCLUSIONS Unlike anti-PD-1 monotherapy, atezolizumab/bevacizumab demonstrated favourable responses in intrahepatic lesions, comparable to those in extrahepatic lesions, and may potentially overcome the immune-tolerant hepatic microenvironment in patients with advanced HCC.
Collapse
Affiliation(s)
- Jaekyung Cheon
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Sanghoon Jung
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jung Sun Kim
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Beodeul Kang
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Hyeyoung Kim
- Department of Hematology-Oncology, Ulsan University Hospital, Ulsan, South Korea
| | - Landon L Chan
- State Key Laboratory of Translational Oncology; Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Department of Clinical Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Lars Becker
- F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Stephen L Chan
- State Key Laboratory of Translational Oncology; Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Department of Clinical Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Chan Kim
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Hong Jae Chon
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| |
Collapse
|
8
|
Saeed A, Tabernero J, Parikh A, Van den Eynde M, Karthaus M, Gerlinger M, Wang Z, Wang G, Smith R, Hecht JR. STELLAR-303: randomized phase III study of zanzalintinib + atezolizumab in previously treated metastatic colorectal cancer. Future Oncol 2024; 20:1733-1743. [PMID: 39041200 PMCID: PMC11485978 DOI: 10.1080/14796694.2024.2352276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/03/2024] [Indexed: 07/24/2024] Open
Abstract
Most patients with metastatic colorectal cancer (mCRC) have limited treatment options following standard-of-care therapy. VEGFR-tyrosine kinase inhibitors (TKIs) have demonstrated clinical activity in mCRC in combination with immune checkpoint inhibitors (ICIs), particularly in patients without liver metastases. The TKI zanzalintinib (XL092) targets VEGFR, MET and TAM kinases, proteins that are involved in tumor growth, angiogenesis, metastasis and immunosuppression. Zanzalintinib has immunomodulatory properties that may enhance response to ICIs. Presented is the design of STELLAR-303, a global, phase III, open-label, randomized study evaluating zanzalintinib plus atezolizumab versus regorafenib in patients with non-MSI-H mCRC who progressed during/after or are refractory/intolerant to standard-of-care therapy. The primary end point is overall survival in patients without liver metastases.Clinical Trial Registration: NCT05425940 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Anwaar Saeed
- University of Pittsburgh Medical Center (UPMC) & UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus & Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Aparna Parikh
- Harvard Medical School, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Marc Van den Eynde
- Institut Roi Albert II, Department of Medical Oncology Cliniques Universitaires St-Luc, Brussels, Belgium
- Institut de Recherche Clinique et Experimentale (Pole MIRO), Université Catholique de Louvain, Brussels, Belgium
| | - Meinolf Karthaus
- Department of Hematology & Oncology, Klinikum Neuperlach/Klinikum Harlaching, Munich, Germany
| | - Marco Gerlinger
- Barts Cancer Institute, Queen Mary University of London, UK
- Gastrointestinal Cancer Centre, St Bartholomew's Hospital, London, UK
| | | | | | | | - J Randolph Hecht
- David Geffen School of Medicine at UCLA, Jonsson Comprehensive Cancer Center, Santa Monica, CA, USA
| |
Collapse
|
9
|
Yang X, Liao Y, Fan L, Lin B, Li J, Wu D, Liao D, Yuan L, Liu J, Gao F, Feng G, Du X. High-intensity focused ultrasound ablation combined with immunotherapy for treating liver metastases: A prospective non-randomized trial. PLoS One 2024; 19:e0306595. [PMID: 38968190 PMCID: PMC11226133 DOI: 10.1371/journal.pone.0306595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
PURPOSE Given the unique features of the liver, it is necessary to combine immunotherapy with other therapies to improve its efficacy in patients of advanced cancer with liver metastases (LM). High-intensity focused ultrasound (HIFU) ablation is now widely used in clinical practice and can enhanced immune benefits. The study is intended to prospectively evaluate the safety and clinical feasibility of HIFU ablation in combination with systemic immunotherapy for patients with liver metastases. METHODS The study enrolled 14 patients with LM who received ultrasound-guided HIFU ablation combined with immune checkpoint inhibitors (ICIs) such as anti-programmed cell death protein 1 (anti-PD-1 agents manufactured in China) at Mianyang Central Hospital. Patients were followed up for adverse events (AEs) during the trial, using the CommonTerminology Criteria for Adverse Events v5.0(CTCAE v5.0) as the standard. Tumour response after treatment was assessed using computerized tomography. RESULTS The 14 patients (age range, 35-84 years) underwent HIFU ablation at 19 metastatic sites and systemic immunotherapy. The mean lesion volume was 179.9 cm3 (maximum: 733.1 cm3). Median follow-up for this trial was 9 months (range: 3-21) months. The study is clinically feasible and acceptable to patients. CONCLUSION This prospective study confirmed that HIFU combined with immunotherapy is clinically feasible and safe for treating liver metastases.
Collapse
Affiliation(s)
- Xiyue Yang
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Yao Liao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Lingli Fan
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Binwei Lin
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Jie Li
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Danfeng Wu
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Dongbiao Liao
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Li Yuan
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Jihui Liu
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Feng Gao
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Gang Feng
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
| | - Xiaobo Du
- Department of Oncology, Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Bardakçi M, Ergun Y. Immunotherapy in gastric cancer with liver metastasis: Challenges and opportunities. World J Gastrointest Surg 2024; 16:1513-1516. [PMID: 38983315 PMCID: PMC11230037 DOI: 10.4240/wjgs.v16.i6.1513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 06/27/2024] Open
Abstract
In this editorial, we review the article by Liu et al published in the World Journal of Gastrointestinal Surgery investigating the efficacy and safety of immunotherapy in patients with gastric cancer (GC) and liver metastasis. GC, the fifth most commonly diagnosed malignancy worldwide, presents a significant challenge due to its multifactorial etiology and a grim prognosis for unresectable or recurrent cases. The advent of immune checkpoint inhibitors (ICIs) has revolutionized oncology; yet liver metastasis has been associated with reduced response rates, progression-free survival, and overall survival in various malignancies. The CheckMate-649 and KEYNOTE-859 trials demonstrated promising results with ICIs in advanced GC, particularly in patients with liver metastasis. However, a meta-analysis of liver metastatic solid tumors revealed worse outcomes with ICIs, highlighting the need for further investigation. While combined therapies, including ICIs with local treatments, show promise in improving outcomes, the nuanced landscape of ICIs in liver metastatic GC necessitates continued research for robust conclusions. The current contradictions in the literature underscore the importance of cautious interpretation and the exploration of tailored approaches to enhance clinical efficacy in this challenging patient population.
Collapse
Affiliation(s)
- Murat Bardakçi
- Department of Medical Oncology, Diyarbakır Gazi Yasargil Training and Research Hospital, Diyarbakir 021000, Kayapinar, Türkiye
| | - Yakup Ergun
- Department of Medical Oncology, Antalya City Hospital, Antalya 07080, Türkiye
| |
Collapse
|
11
|
Zhang X, Wu M, Chen J, Zheng K, Du H, Li B, Gu Y, Jiang J. Comparative efficacy of immune checkpoint inhibitors combined with chemotherapy in patients with advanced driver-gene negative non-small cell lung cancer: A systematic review and network meta-analysis. Heliyon 2024; 10:e30809. [PMID: 38774326 PMCID: PMC11107224 DOI: 10.1016/j.heliyon.2024.e30809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024] Open
Abstract
Objective To evaluate the efficacy of different combinations of immune checkpoint inhibitors (ICIs) and chemotherapy (CT) in the treatment of advanced non-small cell lung cancer (NSCLC). Methods We obtained relevant randomized controlled trials (RCTs) from databases such as PubMed, Embase, Web of Science, and The Cochrane Library up to May 31, 2023. The analysis of clinical prognostic factors was performed using R 4.2.3 and STATA 15.0. The main outcomes measured were overall survival (OS) and progression-free survival (PFS), while secondary outcomes included the objective response rate (ORR), disease control rate (DCR), and treatment-related adverse events of grade 3-5 severity (Grade ≥3 TRAE). Results A total of 17 randomized controlled trials (RCTs) were conducted between 2012 and 2023, involving 7792 patients. These trials evaluated 11 different treatment methods. The results of these trials showed that in terms of overall survival (OS) and progression-free survival (PFS), the combination of tislelizumab with chemotherapy and the combination of camrelizumab with chemotherapy were particularly effective. Moreover, when compared with other combination therapies, pembrolizumab combined with chemotherapy showed superiority in terms of disease control rate (DCR) and objective response rate (ORR). Subgroup analyses further demonstrated that the addition of immune checkpoint inhibitors (ICIs) to chemotherapy significantly improved PFS and OS in patients without liver metastasis and in those with brain metastasis. Additionally, carboplatin-based combination therapy was found to confer favorable survival benefits in terms of PFS, while cisplatin-based combination therapy showed the most favorable outcomes in terms of OS. The results of subgroup analyses for overall survival (OS) showed that the combination of immunotherapy and chemotherapy yielded positive outcomes in specific subgroups. These subgroups were characterized by PD-L1 Tumor Proportion Score (TPS) of 50 % or higher, usage of anti-PD-1 medications, age below 65, male gender, smoking history, and non-squamous cell carcinoma histology. Superior effectiveness was demonstrated only in extending the progression-free survival (PFS) of female patients and patients with squamous carcinoma. Meanwhile, other patient cohorts did not show the same level of improvement. Conclusions Tislelizumab, camrelizumab or pembrolizumab combined with chemotherapy may be the optimal first-line treatment strategies for NSCLC.
Collapse
Affiliation(s)
- Xuewen Zhang
- Department of Oncology, Graduate School of Qinghai University, Qinghai, China
| | - Min Wu
- Department of Oncology, Graduate School of Qinghai University, Qinghai, China
| | - Jie Chen
- Department of Oncology, Graduate School of Qinghai University, Qinghai, China
| | - Kaiman Zheng
- Department of Oncology, Graduate School of Qinghai University, Qinghai, China
| | - Huchen Du
- Department of Oncology, 903 Hosptial, Sichuan, China
| | - Bo Li
- Department of Oncology, Graduate School of Qinghai University, Qinghai, China
| | - Yujia Gu
- Department of Oncology, Graduate School of Qinghai University, Qinghai, China
| | - Jun Jiang
- Division III, Department of Medical Oncology, Affiliated Hospital of Qinghai University, Qinghai, China
| |
Collapse
|
12
|
Korolewicz JA, Scheiner B, Fulgenzi CA, D’Alessio A, Cortellini A, Pascual C, Mehan A, Partridge S, Gujral DM, Mohammed W, Mohammed O, Grzesiak A, Booker L, Cleator S, Pokrovska T, Saleem W, Rackie J, Needham Y, Krell J, McNeish I, Tookman L, Edward Park WH, Asif M, Evans JS, Pinato DJ. The Hammersmith Score Optimises Patient Selection and Predicts for Overall Survival in Early-Phase Cancer Trial Participants Independent of Tumour Burden. Chemotherapy 2024; 69:205-211. [PMID: 38679017 PMCID: PMC11614300 DOI: 10.1159/000539109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION As tumour response rates are increasingly demonstrated in early-phase cancer trials (EPCT), optimal patient selection and accurate prognostication are paramount. Hammersmith Score (HS), a simple prognostic index derived on routine biochemical measures (albumin <35 g/L, lactate dehydrogenase >450 IU/L, sodium <135 mmol/L), is a validated predictor of response and survival in EPCT participants. HS has not been validated in the cancer immunotherapy era. METHODS We retrospectively analysed characteristics and outcomes of unselected referrals to our early-phase unit (12/2019-12/2022). Independent predictors for overall survival (OS) were identified from univariable and multivariable models. HS was calculated for 66 eligible trial participants and compared with the Royal Marsden Score (RMS) to predict OS. Multivariable logistic regression and C-index was used to compare predictive ability of prognostic models. RESULTS Of 212 referrals, 147 patients were screened and 82 patients treated in EPCT. Prognostic stratification by HS identifies significant difference in median OS, and HS was confirmed as a multivariable predictor for OS (HR: HS 1 vs. 0 2.51, 95% CI: 1.01-6.24, p = 0.049; HS 2/3 vs. 0: 10.32, 95% CI: 2.15-49.62, p = 0.004; C-index 0.771) with superior multivariable predictive ability than RMS (HR: RMS 2 vs. 0/1 5.46, 95% CI: 1.12-26.57, p = 0.036; RMS 3 vs. 0/1 6.83, 95% CI: 1.15-40.53, p < 0.001; C-index 0.743). CONCLUSIONS HS is a validated prognostic index for patients with advanced cancer treated in the context of modern EPCTs, independent of tumour burden. HS is a simple, inexpensive prognostic tool to optimise referral for EPCT.
Collapse
Affiliation(s)
| | - Bernhard Scheiner
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Claudia A.M. Fulgenzi
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Department of Medical Oncology, University Campus Bio-Medico of Rome, Roma, Italy
| | - Antonio D’Alessio
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), Novara, Italy
| | - Alessio Cortellini
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Operative Research Unit of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico Roma, Italy
| | - Chynna Pascual
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Aman Mehan
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Sarah Partridge
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Dorothy M. Gujral
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Waleed Mohammed
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Oreoluwa Mohammed
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Aneta Grzesiak
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Lauren Booker
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Susan Cleator
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Tzveta Pokrovska
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Waqar Saleem
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - James Rackie
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Yasmine Needham
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Jonathan Krell
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Iain McNeish
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Laura Tookman
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Won-Ho Edward Park
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Muzamil Asif
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Joanne S. Evans
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - David J. Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), Novara, Italy
| |
Collapse
|
13
|
Kurokami Y, Yamashita C, Yokoi K, Tonomura K, Kiyohara E, Ishitsuka Y, Ono Y, Higashihara H, Fujimoto M, Tanemura A. Successful hepatic arterial chemoembolization for metastatic melanoma resistant to immune checkpoint inhibitors in an extremely aged patient. J Dermatol 2024; 51:e66-e67. [PMID: 37438977 DOI: 10.1111/1346-8138.16894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/10/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023]
Affiliation(s)
- Yu Kurokami
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chigusa Yamashita
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Yokoi
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kyoko Tonomura
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eiji Kiyohara
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yosuke Ishitsuka
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Ono
- Department of Diagnostic and Interventional radiology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroki Higashihara
- Department of High Precision Image-guided Percutaneous Intervention, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
14
|
Stemmer A, Margalit O, Serpas V, Strauss G, Thomas J, Shah P, Tau N, Levanon K, Shacham-Shmueli E, Kopetz S, Overman M, Boursi B. Immunotherapy in mismatch repair-deficient metastatic colorectal cancer - Outcome and novel predictive markers. Eur J Cancer 2024; 198:113495. [PMID: 38157568 DOI: 10.1016/j.ejca.2023.113495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND This study aims to assess predictive markers for response to immunotherapy in dMMR/MSI-H metastatic colorectal cancer (mCRC) patients. MATERIALS AND METHODS A study using two prospective cohorts from MD Anderson Cancer Center and Sheba Medical Center of consecutive patients with dMMR/MSI-H mCRC that were treated with immunotherapy between 2014-2022. Primary outcome was progression-free survival (PFS) and secondary outcome was overall response rate (ORR). Evaluated predictors included ECOG-PS score, RAS/BRAF status, single-agent versus doublet immunotherapy, metastatic sites, disease burden, and CEA levels prior to treatment initiation. Kaplan-Meier analysis and Cox proportional hazard regression model were used to analyze the effect of exposure variables on PFS. RESULTS The study included 153 patients. Median follow-up time was 26 months (IQR 11-48). Median PFS was 51.6 months (95%CI 38.1-NR) and ORR was 58.1%. In a univariate analysis, male sex was associated with worse PFS with a HR of 1.67 (95% CI 1.00-2.79); Right-sided tumors were associated with improved PFS with a HR of 0.56 (95% CI 0.32-0.97); Liver or lung metastasis were associated with worse PFS with HRs of 2.35 (95%CI 1.43-3.88) and 2.30 (95%CI 1.31-4.04), respectively; ECOG-PS score ≥ 2, CEA levels ˃5 μg/L prior to treatment initiation and ≥ 3 metastatic sites were associated with worse PFS with HRs of 2.09 (95%CI 0.98-4.47), 2.23 (95%CI 1.30-3.81) and 3.11 (95%CI 1.61-6.03), respectively. Liver or lung metastasis remained significant in a multivariable model. CONCLUSIONS Extent of disease (worse PFS with high CEA, poor ECOG-PS and ≥3 metastatic sites) and disease location (worse PFS with liver or lung metastasis and left sided tumor) were associated with immunotherapy outcome in dMMR/MSI-H mCRC.
Collapse
Affiliation(s)
- Amos Stemmer
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel.
| | - Ofer Margalit
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel
| | - Victoria Serpas
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gal Strauss
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel
| | - Jane Thomas
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Preksha Shah
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Noam Tau
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Levanon
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel
| | - Einat Shacham-Shmueli
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel
| | - Scott Kopetz
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Overman
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ben Boursi
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Israel; Tel-Aviv University, Tel-Aviv, Israel; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Sanda GE, Shabto JM, Goyal S, Liu Y, Martini DJ, Nazha B, Brown JT, Yantorni LB, Anne Russler G, Caulfield S, Joshi SS, Narayan VM, Kissick H, Ogan K, Master VA, Carthon BC, Kucuk O, Bilen MA. Immune-Related Adverse Events and Clinical Outcomes in Advanced Urothelial Cancer Patients Treated With Immune Checkpoint Inhibitors. Oncologist 2023; 28:1072-1078. [PMID: 37285524 PMCID: PMC10712712 DOI: 10.1093/oncolo/oyad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/26/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND In advanced urothelial cancers (UC), immune checkpoint inhibitors (ICI) show promise as a durable therapy. Immune-related adverse events (irAEs), a side effect of ICIs, may serve as an indicator of beneficial response. We investigated the relationship between irAEs and clinical outcomes in patients with advanced UC who received ICI. MATERIALS AND METHODS In this retrospective study, we investigated 70 patients with advanced UC treated with ICIs at Winship Cancer Institute from 2015 to 2020. Data on patients were collected through chart review. Cox's proportional hazard model and logistic regression were applied to estimate the association with overall survival (OS), progression-free survival (PFS), and clinical benefit (CB). The possible lead-time bias was handled in extended Cox regression models. RESULTS The median age of the cohort was 68. Over one-third (35%) of patients experienced an irAE, with skin being the most frequent organ involved (12.9%). Patients that experienced at least one irAE had significantly enhanced OS (HR: 0.38, 95% CI, 0.18-0.79, P = .009), PFS (HR: 0.27, 95% CI, 0.14-0.53, P < .001), and CB (OR: 4.20, 95% CI, 1.35-13.06, P = .013). Patients who experienced dermatologic irAEs also had significantly greater OS, PFS, and CB. CONCLUSION Of patients with advanced UC that had undergone ICI therapy, those who had irAEs, especially dermatologic irAEs, had significantly greater OS, PFS, and CB. These results may suggest that irAE's may serve as an important marker of durable response to ICI therapy in urothelial cancer. The findings of this study need to be validated with larger cohort studies in the future.
Collapse
Affiliation(s)
- Gregory E Sanda
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Julie M Shabto
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Subir Goyal
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Yuan Liu
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Dylan J Martini
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Bassel Nazha
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Jacqueline T Brown
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Lauren B Yantorni
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Greta Anne Russler
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Sarah Caulfield
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pharmaceutical Services, Emory University School of Medicine, Atlanta, GA, USA
| | - Shreyas S Joshi
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Vikram M Narayan
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Haydn Kissick
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kenneth Ogan
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Viraj A Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bradley C Carthon
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
16
|
Chen EX, Loree JM, Titmuss E, Jonker DJ, Kennecke HF, Berry S, Couture F, Ahmad CE, Goffin JR, Kavan P, Harb M, Colwell B, Samimi S, Samson B, Abbas T, Aucoin N, Aubin F, Koski S, Wei AC, Tu D, O’Callaghan CJ. Liver Metastases and Immune Checkpoint Inhibitor Efficacy in Patients With Refractory Metastatic Colorectal Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2346094. [PMID: 38051531 PMCID: PMC10698621 DOI: 10.1001/jamanetworkopen.2023.46094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/09/2023] [Indexed: 12/07/2023] Open
Abstract
Importance Immune checkpoint inhibitors (ICIs) have limited activity in microsatellite-stable (MSS) or mismatch repair-proficient (pMMR) colorectal cancer. Recent findings suggest the efficacy of ICIs may be modulated by the presence of liver metastases (LM). Objective To investigate the association between the presence of LM and ICI activity in advanced MSS colorectal cancer. Design, Setting, and Participants In this secondary analysis of the Canadian Cancer Trials Group CO26 (CCTG CO.26) randomized clinical trial, patients with treatment-refractory colorectal cancer were randomized in a 2:1 fashion to durvalumab plus tremelimumab or best supportive care alone between August 10, 2016, and June 15, 2017. The primary end point was overall survival (OS) with 80% power and 2-sided α = .10. The median follow-up was 15.2 (0.2-22.0) months. In this post hoc analysis performed from February 11 to 14, 2022, subgroups were defined based on the presence or absence of LM and study treatments. Intervention Durvalumab plus tremelimumab or best supportive care. Main Outcomes and Measures Hazard ratios (HRs) and 90% CIs were calculated based on a stratified Cox proportional hazards regression model. Plasma tumor mutation burden at study entry was determined using a circulating tumor DNA assay. The primary end point of the study was OS, defined as the time from randomization to death due to any cause; secondary end points included progression-free survival (PFS) and disease control rate (DCR). Results Of 180 patients enrolled (median age, 65 [IQR, 36-87] years; 121 [67.2%] men; 19 [10.6%] Asian, 151 [83.9%] White, and 10 [5.6%] other race or ethnicity), LM were present in 127 (70.6%). For patients with LM, there was a higher proportion of male patients (94 of 127 [74.0%] vs 27 of 53 [50.9%]; P = .005), and the time from initial cancer diagnosis to study entry was shorter (median, 40 [range, 8-153] vs 56 [range, 14-181] months; P = .001). Plasma tumor mutation burden was significantly higher in patients with LM. Patients without LM had significantly improved PFS with durvalumab plus tremelimumab (HR, 0.54 [90% CI, 0.35-0.96]; P = .08; P = .02 for interaction). Disease control rate was 49% (90% CI, 36%-62%) in patients without LM treated with durvalumab plus tremelimumab, compared with 14% (90% CI, 6%-38%) in those with LM (odds ratio, 5.70 [90% CI, 1.46-22.25]; P = .03). On multivariable analysis, patients without LM had significantly improved OS and PFS compared with patients with LM. Conclusions and Relevance In this secondary analysis of the CCTG CO.26 study, the presence of LM was associated with worse outcomes for patients with advanced colorectal cancer. Patients without LM had improved PFS and higher DCR with durvalumab plus tremelimumab. Liver metastases may be associated with poor outcomes of ICI treatment in advanced colorectal cancer and should be considered in the design and interpretation of future clinical studies evaluating this therapy.
Collapse
Affiliation(s)
- Eric X. Chen
- Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | | | - Emma Titmuss
- British Columbia Cancer Agency, Vancouver, Canada
| | - Derek J. Jonker
- The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Hagen F. Kennecke
- Portland Providence Cancer Center, Earle Chiles Research Institute, Portland, Oregon
| | - Scott Berry
- Department of Oncology, Queen’s University, Kingston, Ontario, Canada
| | | | | | | | - Petr Kavan
- Segal Cancer Center, Montreal, Quebec, Canada
| | | | - Bruce Colwell
- Queen Elizabeth II Health Sciences Center, Halifax, Nova Scotia, Canada
| | - Setareh Samimi
- Hôpital Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Benoit Samson
- Charles LeMoyne Hospital Cancer Centre, Sherbrooke, Quebec, Canada
| | - Tahir Abbas
- Saskatoon Cancer Center, Saskatoon, Saskatoon, Canada
| | | | - Francine Aubin
- Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | | | - Alice C. Wei
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | | |
Collapse
|
17
|
Yu H, Chen J, Zhou Y, Sheng J, Zhang X, He L, Chen L, Chu Q, Zhang L, Hong S. Impact of prophylactic dexamethasone on the efficacy of immune checkpoint inhibitors plus platinum-based chemotherapy in patients with advanced Non-Squamous Non-Small-Cell lung cancer. Int Immunopharmacol 2023; 125:111138. [PMID: 37948858 DOI: 10.1016/j.intimp.2023.111138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Baseline corticosteroids exposure is associated with inferior clinical outcomes in patients with non-small-cell lung cancer (NSCLC) treated with programmed cell death-1 (PD-1) axis blockade. Dexamethasone is a potent corticosteroid used in the prevention of chemotherapy-associated adverse events (CAAEs). OBJECTIVE Since dexamethasone has immunosuppressive properties, this study attempted to elucidate its effects on the efficacy of immunotherapy plus chemotherapy in patients with non-squamous NSCLC. METHODS The study retrospectively analyzed the medical records of 254 advanced non-squamous NSCLC patients who received front-line treatment with a PD-1 pathway inhibitor and platinum-based chemotherapy at three academic institutions. The average dosage of prophylactic dexamethasone per chemotherapy cycle was calculated. Patients were divided into three groups based on the dose of dexamethasone: High-d (≥24 mg), Moderate-d (12-24 mg), and Low-d (<12 mg). Spearman's rank correlation was used to assess the correlation between the dosage of dexamethasone and progression-free survival (PFS). Logistic regression was used to assess the correlation between dexamethasone dosage and the occurrence of immune related adverse effects (irAE). Univariate and multivariate Cox proportional hazards regression models were used to analyze the differences in survival among the different dexamethasone dosage groups. RESULT The dosage of prophylactic dexamethasone was not significantly correlated with PFS (Spearman's rho = -0.103, P = 0.098). Results from the univariate [hazard ratio (HR)Low-d/High-d, 1.00; P = 0.997; HRModerate-d/High-d, 0.85; P = 0.438] and multivariate (HRLow-d/High-d, 0.71; P = 0.174; HRModerate-d/High-d, 0.87; P = 0.512) analyses showed no significant association between dexamethasone and PFS. Dexamethasone did not have significant effect on the objective response rate, disease control rate or overall survival. The toxicity profiles of irAE were similar across all three groups. CONCLUSION The results of this study suggest that the use of prophylactic dexamethasone does not have an adverse effect on the clinical outcomes of non-squamous NSCLC patients treated with PD-1 blockade therapy and chemotherapy. Routine use of dexamethasone for preventing CAAEs should be recommended for patients undergoing combined immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Hui Yu
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jingyu Chen
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yixin Zhou
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jin Sheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang 310018, Hangzhou, China
| | - Xuanye Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Lina He
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Likun Chen
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| |
Collapse
|
18
|
Margalit O, Lieberman S, Redinsky I, Halparin S, Honig N, Raskin S, Ben-Ayun M, Shacham-Shmueli E, Halpern N, Urban D, Ackerstein A, Shulman K, Ben-Ami E, Semenisty V, Purim O, Yarom N, Golan T, Boursi B, Appel S, Symon Z, Berger R, Mauro D, Krieg AM, Lawrence YR. Combination Treatment of Intratumoral Vidutolimod, Radiosurgery, Nivolumab, and Ipilimumab for Microsatellite Stable Colorectal Carcinoma With Liver Metastases. Clin Colorectal Cancer 2023; 22:442-449.e1. [PMID: 37657954 DOI: 10.1016/j.clcc.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 09/03/2023]
Abstract
INTRODUCTION Microsatellite stable metastatic colorectal cancer (MSS mCRC) is largely refractory to immune checkpoint inhibition. We hypothesized that a combination of intratumoral TLR9 agonist, radiosurgery and dual PD-1 and CTLA-4 blockade would induce a local focus of immune stimulation, evoking a systemic immune response. PATIENTS AND METHODS In this phase I single-institution study, patients with MSS mCRC were treated with a priming dose of s.c vidutolimod, 3 intratumoral injections of vidutolimod and radiosurgery, combined with nivolumab and ipilimumab. Cytokine levels were measured at baseline and at 7 (± 2) weeks. Patients were accrued to 4 consecutive cohorts: (1) Safety run-in without radiosurgery, (2) Radiosurgery prior to intratumoral therapy, (3) Radiosurgery prior to intratumoral therapy with a condensed timeline, and (4) Radiosurgery to extrahepatic lesion following completion of intratumoral therapy. RESULTS A total of 19 patients were accrued. Median age was 59 years (range 40-71), 68% were male, median number of previous systemic treatments was 3 (range 2-5). None of the patients responded, aside from 1 patient, attributed to high tumor mutational burden. Grade 3 liver toxicity was reported in 0%, 0%, 75%, and 17% in cohorts 1 to 4, respectively. Systemic levels of CXCL10 and IL-10 increased, with a median of 407 versus 78 pg/mL (P = .01), and 66 versus 40 pg/mL (P = .03), respectively. CONCLUSIONS The combination of intratumoral vidutolimod, radiosurgery, nivolumab and ipilimumab was not found to be efficacious in MSS mCRC with liver metastases. The juxtaposition of liver irradiation and intratumoral vidutolimod injection was associated with high hepatic toxicity.
Collapse
Affiliation(s)
- Ofer Margalit
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Sivan Lieberman
- Department of Diagnostic Imaging, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Ilanit Redinsky
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Sharon Halparin
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Nir Honig
- Department of Radiation Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Stephen Raskin
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel; Department of Diagnostic Imaging, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Maoz Ben-Ayun
- Department of Radiation Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Einat Shacham-Shmueli
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Naama Halpern
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Damien Urban
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Aliza Ackerstein
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Katerina Shulman
- Department of Medical Oncology, Lady Davis Carmel Hospital, Haifa, Israel
| | - Eytan Ben-Ami
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Valeriya Semenisty
- Department of Medical Oncology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Ofer Purim
- Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nirit Yarom
- Department of Medical Oncology, Shamir Medical Center, Beer Yaacov, Israel
| | - Talia Golan
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Ben Boursi
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Sarit Appel
- Department of Radiation Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Zvi Symon
- Department of Radiation Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Raanan Berger
- Department of Medical Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel
| | | | | | - Yaacov R Lawrence
- Department of Radiation Oncology, Sidney Kimmel Medical College & Cancer Center at Thomas Jefferson University-Jefferson Health, Phila, PA; Department of Radiation Oncology, Sheba Medical Center, Ramat Gan affiliated with Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
19
|
Zlotnik O, Krzywon L, Bloom J, Kalil J, Altubi I, Lazaris A, Metrakos P. Targeting Liver Metastases to Potentiate Immunotherapy in MS-Stable Colorectal Cancer-A Review of the Literature. Cancers (Basel) 2023; 15:5210. [PMID: 37958384 PMCID: PMC10649257 DOI: 10.3390/cancers15215210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Immunotherapy has revolutionized the treatment of several cancers, including melanoma and lung cancer. However, for colorectal cancer, it is ineffective for 95% of patients with microsatellite-stable disease. Recent evidence suggests that the liver's immune microenvironment plays a pivotal role in limiting the effectiveness of immunotherapy. There is also evidence to show that targeting liver metastases with locoregional therapies, such as surgery or irradiation, could potentiate immunotherapy for these patients. This review presents evidence from preclinical studies regarding the underlying mechanisms and from clinical studies that support this approach. Furthermore, we outline potential directions for future clinical trials. This innovative strategy could potentially establish immunotherapy as an effective treatment for MS-stable colorectal cancer patients, which are currently considered resistant.
Collapse
Affiliation(s)
- Oran Zlotnik
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (O.Z.); (L.K.); (J.B.); (J.K.); (A.L.)
- Division of General Surgery, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Lucyna Krzywon
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (O.Z.); (L.K.); (J.B.); (J.K.); (A.L.)
| | - Jessica Bloom
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (O.Z.); (L.K.); (J.B.); (J.K.); (A.L.)
| | - Jennifer Kalil
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (O.Z.); (L.K.); (J.B.); (J.K.); (A.L.)
- Division of General Surgery, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Ikhtiyar Altubi
- Division of General Surgery, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Anthoula Lazaris
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (O.Z.); (L.K.); (J.B.); (J.K.); (A.L.)
| | - Peter Metrakos
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (O.Z.); (L.K.); (J.B.); (J.K.); (A.L.)
- Division of General Surgery, McGill University, Montreal, QC H4A 3J1, Canada;
| |
Collapse
|
20
|
Hernando-Calvo A, Vila-Casadesús M, Bareche Y, Gonzalez-Medina A, Abbas-Aghababazadeh F, Lo Giacco D, Martin A, Saavedra O, Brana I, Vieito M, Fasani R, Stagg J, Mancuso F, Haibe-Kains B, Han M, Berche R, Pugh TJ, Mirallas O, Jimenez J, Gonzalez NS, Valverde C, Muñoz-Couselo E, Suarez C, Diez M, Élez E, Capdevila J, Oaknin A, Saura C, Macarulla T, Galceran JC, Felip E, Dienstmann R, Bedard PL, Nuciforo P, Seoane J, Tabernero J, Garralda E, Vivancos A. A pan-cancer clinical platform to predict immunotherapy outcomes and prioritize immuno-oncology combinations in early-phase trials. MED 2023; 4:710-727.e5. [PMID: 37572657 DOI: 10.1016/j.medj.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 06/01/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Immunotherapy is effective, but current biomarkers for patient selection have proven modest sensitivity. Here, we developed VIGex, an optimized gene signature based on the expression level of 12 genes involved in immune response with RNA sequencing. METHODS We implemented VIGex using the nCounter platform (Nanostring) on a large clinical cohort encompassing 909 tumor samples across 45 tumor types. VIGex was developed as a continuous variable, with cutoffs selected to detect three main categories (hot, intermediate-cold and cold) based on the different inflammatory status of the tumor microenvironment. FINDINGS Hot tumors had the highest VIGex scores and exhibited an increased abundance of tumor-infiltrating lymphocytes as compared with the intermediate-cold and cold. VIGex scores varied depending on tumor origin and anatomic site of metastases, with liver metastases showing an immunosuppressive tumor microenvironment. The predictive power of VIGex-Hot was observed in a cohort of 98 refractory solid tumor from patients treated in early-phase immunotherapy trials and its clinical performance was confirmed through an extensive metanalysis across 13 clinically annotated gene expression datasets from 877 patients treated with immunotherapy agents. Last, we generated a pan-cancer biomarker platform that integrates VIGex categories with the expression levels of immunotherapy targets under development in early-phase clinical trials. CONCLUSIONS Our results support the clinical utility of VIGex as a tool to aid clinicians for patient selection and personalized immunotherapy interventions. FUNDING BBVA Foundation; 202-2021 Division of Medical Oncology and Hematology Fellowship award; Princess Margaret Cancer Center.
Collapse
Affiliation(s)
- Alberto Hernando-Calvo
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain; Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C4, Canada; Departamento de Medicina, Universidad Autónoma de Barcelona (UAB), 08035 Barcelona, Spain
| | | | - Yacine Bareche
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0A9, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, QC H3T1J4, Canada
| | | | | | | | - Agatha Martin
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Omar Saavedra
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Irene Brana
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Maria Vieito
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Roberta Fasani
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - John Stagg
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X0A9, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, QC H3T1J4, Canada
| | | | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G2C4, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G1L7, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5S2E4, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G0A3, Canada; Vector Institute for Artificial Intelligence, Toronto, ON M5G1M1, Canada
| | - Ming Han
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G2C4, Canada
| | - Roger Berche
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G2C4, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G1L7, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G0A3, Canada
| | - Oriol Mirallas
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Jose Jimenez
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Nadia Saoudi Gonzalez
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Claudia Valverde
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Eva Muñoz-Couselo
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Cristina Suarez
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Marc Diez
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Elena Élez
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Jaume Capdevila
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Ana Oaknin
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Cristina Saura
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Teresa Macarulla
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Joan Carles Galceran
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Enriqueta Felip
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | | | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C4, Canada
| | - Paolo Nuciforo
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Josep Tabernero
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Elena Garralda
- Department of Medical Oncology, Vall D'Hebron University Hospital, 08035 Barcelona, Spain; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Ana Vivancos
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain.
| |
Collapse
|
21
|
Foote MB, Argilés G, Rousseau B, Segal NH. Facts and Hopes in Colorectal Cancer Immunotherapy. Clin Cancer Res 2023; 29:4032-4039. [PMID: 37326624 DOI: 10.1158/1078-0432.ccr-22-2176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Although a minority of colorectal cancers exhibit mismatch repair deficiency and associated sensitivity to immune checkpoint inhibitors (ICI), the vast majority of colorectal cancers arise in a tolerogenic microenvironment with mismatch repair proficiency, low tumor-intrinsic immunogenicity, and negligible immunotherapy responsiveness. Treatment strategies to augment tumor immunity with combination ICIs and chemotherapy have broadly failed in mismatch repair-proficient tumors. Similarly, although several small single-arm studies have shown that checkpoint blockade plus radiation or select tyrosine kinase inhibition may show improved outcomes compared with historical controls, this finding has not been clearly validated in randomized trials. An evolving next generation of intelligently engineered checkpoint inhibitors, bispecific T-cell engagers, and emerging CAR-T cell therapies may improve immunorecognition of colorectal tumors. Across these modalities, ongoing translational efforts to better define patient populations and biomarkers associated with immune response, as well as combine biologically sound and mutually amplifying therapies, show promise for a new era of immunotherapy in colorectal cancer.
Collapse
Affiliation(s)
- Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Guillem Argilés
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benoit Rousseau
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neil H Segal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
22
|
Patel RK, Rahman S, Schwantes IR, Bartlett A, Eil R, Farsad K, Fowler K, Goodyear SM, Hansen L, Kardosh A, Nabavizadeh N, Rocha FG, Tsikitis VL, Wong MH, Mayo SC. Updated Management of Colorectal Cancer Liver Metastases: Scientific Advances Driving Modern Therapeutic Innovations. Cell Mol Gastroenterol Hepatol 2023; 16:881-894. [PMID: 37678799 PMCID: PMC10598050 DOI: 10.1016/j.jcmgh.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths in the United States and accounts for an estimated 1 million deaths annually worldwide. The liver is the most common site of metastatic spread from colorectal cancer, significantly driving both morbidity and mortality. Although remarkable advances have been made in recent years in the management for patients with colorectal cancer liver metastases, significant challenges remain in early detection, prevention of progression and recurrence, and in the development of more effective therapeutics. In 2017, our group held a multidisciplinary state-of-the-science symposium to discuss the rapidly evolving clinical and scientific advances in the field of colorectal liver metastases, including novel early detection and prognostic liquid biomarkers, identification of high-risk cohorts, advances in tumor-immune therapy, and different regional and systemic therapeutic strategies. Since that time, there have been scientific discoveries translating into therapeutic innovations addressing the current management challenges. These innovations are currently reshaping the treatment paradigms and spurring further scientific discovery. Herein, we present an updated discussion of both the scientific and clinical advances and future directions in the management of colorectal liver metastases, including adoptive T-cell therapies, novel blood-based biomarkers, and the role of the tumor microbiome. In addition, we provide a comprehensive overview detailing the role of modern multidisciplinary clinical approaches used in the management of patients with colorectal liver metastases, including considerations toward specific molecular tumor profiles identified on next generation sequencing, as well as quality of life implications for these innovative treatments.
Collapse
Affiliation(s)
- Ranish K Patel
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Shahrose Rahman
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Issac R Schwantes
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Alexandra Bartlett
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon
| | - Robert Eil
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon; The Knight Cancer Institute, OHSU, Portland, Oregon
| | - Khashayar Farsad
- Charles T. Dotter Department of Interventional Radiology, OHSU, Portland, Oregon
| | - Kathryn Fowler
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Shaun M Goodyear
- The Knight Cancer Institute, OHSU, Portland, Oregon; Division of Hematology and Oncology, School of Medicine, OHSU, Portland, Oregon
| | - Lissi Hansen
- The Knight Cancer Institute, OHSU, Portland, Oregon; School of Nursing, OHSU, Portland, Oregon
| | - Adel Kardosh
- The Knight Cancer Institute, OHSU, Portland, Oregon; Division of Hematology and Oncology, School of Medicine, OHSU, Portland, Oregon
| | - Nima Nabavizadeh
- The Knight Cancer Institute, OHSU, Portland, Oregon; Department of Radiation Medicine, OHSU, Portland, Oregon
| | - Flavio G Rocha
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon; The Knight Cancer Institute, OHSU, Portland, Oregon
| | - V Liana Tsikitis
- The Knight Cancer Institute, OHSU, Portland, Oregon; Division of Gastrointestinal Surgery, Department of Surgery, OHSU, Portland, Oregon
| | - Melissa H Wong
- The Knight Cancer Institute, OHSU, Portland, Oregon; Department of Cell, Developmental and Cancer Biology, OHSU, Portland, Oregon
| | - Skye C Mayo
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon; The Knight Cancer Institute, OHSU, Portland, Oregon.
| |
Collapse
|
23
|
Waninger JJ, Ma VT, Chopra Z, Pearson AN, Green MD. Evaluation of the Prognostic Role of Liver Metastases on Patient Outcomes: Systematic Review and Meta-analysis. Cancer J 2023; 29:279-284. [PMID: 37796646 PMCID: PMC10558088 DOI: 10.1097/ppo.0000000000000683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
ABSTRACT The liver is a common site of metastasis for many primary malignancies, but the quantitative impact on survival is unknown. We performed a systematic review and meta-analysis of 83 studies (604,853 patients) assessing the overall hazard associated with liver metastases by primary tumor type and treatment regimen. The pooled overall survival hazard ratio (HR) for all included studies was 1.77 (95% confidence interval [CI], 1.62-1.93). Patients with breast cancer primaries fared the worst (HR, 2.37; 95% CI, 1.64-3.44), as did patients treated with immunotherapies (HR, 1.86; 95% CI, 1.42-2.42). Liver metastases negatively impact survival, necessitating new approaches to disease management.
Collapse
Affiliation(s)
| | - Vincent T. Ma
- Department of Internal Medicine, Division of Hematology, Medical Oncology, and Palliative Care, University of Wisconsin, Madison, WI
- Department of Internal Medicine, Division of Hematology and Medical Oncology, University of Michigan, Ann Arbor, MI
| | - Zoey Chopra
- Department of Economics, University of Michigan, Ann Arbor, MI
| | - Ashley N. Pearson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Michael D. Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48109, USA
| |
Collapse
|
24
|
Lin Y, Luo S, Luo M, Lu X, Li Q, Xie M, Huang Y, Liao X, Zhang Y, Li Y, Liang R. Homologous recombination repair gene mutations in colorectal cancer favors treatment of immune checkpoint inhibitors. Mol Carcinog 2023; 62:1271-1283. [PMID: 37232365 DOI: 10.1002/mc.23562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023]
Abstract
Immune checkpoint inhibitor (ICI) therapy is insensitive for Colorectal cancer (CRC) patients with microsatellite stable (MSS). Genomic data of three CRC cohort, n = 35), and the Cancer Genome Atlas (TCGA CRC cohort, n = 377), were analyzed. A cohort treated with ICIs from Memorial Sloan Kettering Cancer Center (MSKCC CRC cohort, n = 110) and two cases from the local hospital were characterized the impact of the HRR mutation on prognosis of CRC. Homologous recombination repair (HRR) gene mutations were more common in CN and HL cohorts (27.85%; 48.57%) than in TCGA CRC cohort (15.92%), especially in the MSS populations, the frequencies of HRR mutation were higher in CN and HL cohort (27.45%, 51.72%) than in TCGA cohort (6.85%). HRR mutations were associated with high tumor mutational burden (TMB-H). Although HRR mutation uncorrelated with an improved overall survival in the MSKCC CRC cohort (p = 0.97), HRR mutated patients had a significantly improved OS compared to the HRR wildtype population particularly in MSS subgroups (p = 0.0407) under ICI treatment. It probably contributed by a higher neoantigen and increased CD4+ T cell infiltration which found in the TCGA MSS HRR mutated CRC cohort. The similar phenomenon on cases was observed that MSS metastatic CRC patient with HRR mutation seemed more sensitive to ICI after multi-line chemotherapy in clinical practice than HRR wildtype. This finding suggests the feasibility of HRR mutation as an immunotherapy response predictor in MSS CRC, which highlights a potential therapeutic approach for these patients.
Collapse
Affiliation(s)
- Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Shanshan Luo
- Department of Gastrointestinal Gland Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Min Luo
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Xuerou Lu
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Qian Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Mingzhi Xie
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Yu Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Xiaoli Liao
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Yumei Zhang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Yongqiang Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
25
|
Tian BW, Han CL, Wang HC, Yan LJ, Ding ZN, Liu H, Mao XC, Tian JC, Xue JS, Yang LS, Tan SY, Dong ZR, Yan YC, Wang DX, Li T. Effect of liver metastasis on the efficacy of immune checkpoint inhibitors in cancer patients: a systemic review and meta-analysis. Clin Exp Metastasis 2023; 40:255-287. [PMID: 37308706 DOI: 10.1007/s10585-023-10217-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2024]
Abstract
Liver metastasis is a frequent phenomenon in advanced tumor disease. Immune checkpoint inhibitors (ICIs) are a new class of therapeutics that can improve the prognosis of cancer patients. The purpose of this study is to elucidate the relationship between liver metastasis and survival outcomes of patients receiving ICIs treatment. We searched four main databases, including PubMed, EMBASE, Cochrane Library, and Web of Science. Overall survival (OS) and progression-free survival (PFS) were the survival outcomes of our concern. Hazard ratio (HR) with 95% confidence interval (CI) were used to evaluate the relationship between liver metastasis and OS/ PFS. Finally, 163 articles were included in the study. The pooled results showed that patients with liver metastasis receiving ICIs treatment had worse OS (HR=1.82, 95%CI:1.59-2.08) and PFS (HR=1.68, 95%CI:1.49-1.89) than patients without liver metastasis. The effect of liver metastasis on ICIs efficacy differed in different tumor types, and patients with urinary system tumors (renal cell carcinoma OS: HR=2.47, 95%CI:1.76-3.45; urothelial carcinoma OS: HR=2.37, 95%CI:2.03-2.76) had the worst prognosis, followed by patients with melanoma (OS: HR=2.04, 95%CI:1.68-2.49) or non-small cell lung cancer (OS: HR=1.81, 95%CI:1.72-1.91). ICIs efficacy in digestive system tumors (colorectal cancer OS: HR=1.35, 95%CI:1.07-1.71; gastric cancer/ esophagogastric cancer OS: HR=1.17, 95%CI:0.90-1.52) was less affected, and peritoneal metastasis and the number of metastases have a greater clinical significance than liver metastasis based on univariate data. For cancer patients receiving ICIs treatment, the occurrence of liver metastasis is associated with poor prognosis. Different cancer types and metastatic sites may hold a different prognostic effect on the efficacy of ICIs treatment in cancer patients.
Collapse
Affiliation(s)
- Bao-Wen Tian
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Cheng-Long Han
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Han-Chao Wang
- Institute for Financial Studies, Shandong Univeristy, Jinan, 250100, People's Republic of China
| | - Lun-Jie Yan
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Zi-Niu Ding
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Hui Liu
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Xin-Cheng Mao
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Jin-Cheng Tian
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Jun-Shuai Xue
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Long-Shan Yang
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Si-Yu Tan
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Zhao-Ru Dong
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Yu-Chuan Yan
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Dong-Xu Wang
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China
| | - Tao Li
- Department of general surgery, Qilu Hospital, Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
26
|
Liang L, Li Y, Hong Y, Ji T, Chen H, Lin Z. Nomogram Based on Liver Function Test Indicators for Survival Prediction in Nasopharyngeal Carcinoma Patients Receiving PD-1 Inhibitor Therapy. Curr Oncol 2023; 30:7189-7202. [PMID: 37623002 PMCID: PMC10453561 DOI: 10.3390/curroncol30080521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 08/26/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the prognostic significance of PD-1 inhibitor therapy in nasopharyngeal carcinoma (NPC) and to develop a nomogram to estimate individual risks. METHODS We retrospectively analyzed 162 NPC patients who were administered the PD-1 inhibitor combined with radiotherapy and chemotherapy at the Sun Yat-Sen University Cancer Center. In total, 108 NPC patients were included in the training cohort and 54 NPC patients were included in the validation cohort. Univariate and multivariate Cox survival analyses were performed to determine the prognostic factors for 1-year and 2-year progression-free survival (PFS). In addition, a nomogram model was constructed to predict the survival probability of PFS. A consistency index (C-index), a decision curve, a clinical impact curve, and a standard curve were used to measure predictive accuracy, the clinical net benefit, and the consistency of prognostic factors. RESULTS Univariate and multivariate analyses indicated that the metastasis stage, the levels of ALT, the AST/ALT ratio, and the LDH were independent risk factors associated with the prognosis of PD-1 inhibitor therapy. A nomogram based on these four indicators was constructed and the Kaplan-Meier survival analysis showed that patients with a higher total score have a shorter PFS. The C-index of this model was 0.732 in the training cohort and 0.847 in the validation cohort, which are higher than those for the TNM stages (training cohort: 0.617; validation cohort: 0.727; p <0.05). Decision Curve Analysis (DCA), Net Reclassification Improvement (NRI), and Integrated Discrimination Improvement (IDI) showed that our model has better prediction accuracy than TNM staging. CONCLUSIONS Predicting PFS in NPC patients based on liver function-related indicators before PD-1 treatment may help clinicians predict the efficacy of PD-1 treatment in these patients.
Collapse
Affiliation(s)
- Lixia Liang
- Department of Clinical Laboratory, The First People’s Hospital of Zhaoqing, Zhaoqing 526060, China; (L.L.); (Y.L.); (Y.H.)
| | - Yan Li
- Department of Clinical Laboratory, The First People’s Hospital of Zhaoqing, Zhaoqing 526060, China; (L.L.); (Y.L.); (Y.H.)
| | - Yansui Hong
- Department of Clinical Laboratory, The First People’s Hospital of Zhaoqing, Zhaoqing 526060, China; (L.L.); (Y.L.); (Y.H.)
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510060, China;
| | - Hao Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Zhifang Lin
- Department of Clinical Laboratory, The First People’s Hospital of Zhaoqing, Zhaoqing 526060, China; (L.L.); (Y.L.); (Y.H.)
| |
Collapse
|
27
|
Han CL, Tian BW, Yan LJ, Ding ZN, Liu H, Mao XC, Tian JC, Xue JS, Tan SY, Dong ZR, Yan YC, Hong JG, Chen ZQ, Wang DX, Li T. Efficacy and safety of immune checkpoint inhibitors for hepatocellular carcinoma patients with macrovascular invasion or extrahepatic spread: a systematic review and meta-analysis of 54 studies with 6187 hepatocellular carcinoma patients. Cancer Immunol Immunother 2023; 72:1957-1969. [PMID: 36811662 PMCID: PMC10991272 DOI: 10.1007/s00262-023-03390-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/27/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND AND AIMS The impacts of macrovascular invasion (MVI) or extrahepatic spread (EHS) on the efficacy and safety of immune checkpoint inhibitors (ICIs) among hepatocellular carcinoma (HCC) patients remain unclear. Thus, we conducted a systematic review and meta-analysis to clarify whether ICI therapy is a feasible treatment option for HCC with MVI or EHS. METHODS Eligible studies published before September 14, 2022, were retrieved. In this meta-analysis, the objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and occurrence of adverse events (AEs) were outcomes of interest. RESULTS Fifty-four studies involving 6187 individuals were included. The findings indicated that the presence of EHS in ICI-treated HCC patients may indicate an inferior ORR (OR 0.77, 95% CI 0.63-0.96), but may not significantly affect the PFS (multivariate analyses: HR 1.27, 95% CI 0.70-2.31) and OS (multivariate analyses: HR 1.23, 95% CI 0.70-2.16). Additionally, the presence of MVI in ICI-treated HCC patients may not have significant prognostic impact on ORR (OR 0.84, 95% CI 0.64-1.10), but may indicate inferior PFS (multivariate analyses: HR 1.75, 95% CI 1.07-2.84) and OS (multivariate analyses: HR 2.03, 95% CI 1.31-3.14). The presence of EHS or MVI in ICI-treated HCC patients may not significantly impact the occurrence of any serious immune-related adverse events (irAEs) (grades ≥ 3) (EHS: OR 0.44, 95% CI 0.12-1.56; MVI: OR 0.68, 95% CI 0.24-1.88). CONCLUSION The presence of MVI or EHS in ICI-treated HCC patients may not significantly impact the occurrence of serious irAEs. However, the presence of MVI (but not EHS) in ICI-treated HCC patients may be a significant negative prognostic factor. Therefore, ICI-treated HCC patients with MVI warrant more attention.
Collapse
Affiliation(s)
- Cheng-Long Han
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Bao-Wen Tian
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Lun-Jie Yan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zi-Niu Ding
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Hui Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Xin-Cheng Mao
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Jin-Cheng Tian
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Jun-Shuai Xue
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Si-Yu Tan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Yu-Chuan Yan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Jian-Guo Hong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zhi-Qiang Chen
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Dong-Xu Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, The Second Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
28
|
Tsilimigras DI, Ntanasis-Stathopoulos I, Pawlik TM. Molecular Mechanisms of Colorectal Liver Metastases. Cells 2023; 12:1657. [PMID: 37371127 DOI: 10.3390/cells12121657] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The liver is the most frequently target for metastasis among patients with colorectal cancer mainly because of the portal vein circulation that directly connects the colon and rectum with the liver. The liver tumor microenvironment consists of different cell types each with unique characteristics and functions that modulate the antigen recognition and immune system activation. Primary tumors from other sites "prime" the liver prior to the seeding of cancer cells, creating a pre-metastatic niche. Following invasion into the liver, four different phases are key to the development of liver metastases: a microvascular phase in which cancer cells infiltrate and become trapped in sinusoidal vessels; an extravascular, pre-angiogenic phase; an angiogenic phase that supplies oxygen and nutrients to cancer cells; and a growth phase in which metastatic cells multiply and enlarge to form detectable tumors. Exosomes carry proteins, lipids, as well as genetic information that can create a pre-metastatic niche in distant sites, including the liver. The complexity of angiogenic mechanisms and the exploitation of the vasculature in situ by cancer cells have limited the efficacy of currently available anti-angiogenic therapies. Delineating the molecular mechanisms implicated in colorectal liver metastases is crucial to understand and predict tumor progression; the development of distant metastases; and resistance to chemotherapy, immunotherapy, and targeted treatment.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Columbus, OH 43210, USA
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Columbus, OH 43210, USA
| |
Collapse
|
29
|
Huang KS, Huang YH, Chen CT, Chou CP, Pan BL, Lee CH. Liver-specific metastases as an independent prognostic factor in cancer patients receiving hospice care in hospital. BMC Palliat Care 2023; 22:62. [PMID: 37221588 DOI: 10.1186/s12904-023-01180-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Survival prediction is important in cancer patients receiving hospice care. Palliative prognostic index (PPI) and palliative prognostic (PaP) scores have been used to predict survival in cancer patients. However, cancer primary site with metastatic status, enteral feeding tubes, Foley catheter, tracheostomy, and treatment interventions are not considered in aforementioned tools. The study aimed to investigate the cancer features and potential clinical factors other than PPI and PaP to predict patient survival. METHODS We conducted a retrospective study for cancer patients admitted to a hospice ward between January 2021 and December 2021. We examined the correlation of PPI and PaP scores with survival time since hospice ward admission. Multiple linear regression was used to test the potential clinical factors other than PPI and PaP for predicting survival. RESULTS A total of 160 patients were enrolled. The correlation coefficients for PPI and PaP scores with survival time were -0.305 and -0.352 (both p < 0.001), but the predictabilities were only marginal at 0.087 and 0.118, respectively. In multiple regression, liver metastasis was an independent poor prognostic factor as adjusted by PPI (β = -8.495, p = 0.013) or PaP score (β = -7.139, p = 0.034), while feeding gastrostomy or jejunostomy were found to prolong survival as adjusted by PPI (β = 24.461, p < 0.001) or PaP score (β = 27.419, p < 0.001). CONCLUSIONS Association between PPI and PaP with patient survival in cancer patients at their terminal stages is low. The presence of liver metastases is a poor survival factor independent of PPI and PaP score.
Collapse
Affiliation(s)
- Kun-Siang Huang
- Department of Family Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yun-Hwa Huang
- Department of Family Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chao-Tung Chen
- Department of Family Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chia-Pei Chou
- Department of Family Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Bo-Lin Pan
- Department of Family Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chih-Hung Lee
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
- Chang Gung University College of Medicine, Taoyuan, Taiwan.
- Institute of Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
- National Sun Yat-Sen University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
30
|
Pierrard J, Van Ooteghem G, Van den Eynde M. Implications of the Organ-Specific Immune Environment for Immune Priming Effect of Radiotherapy in Metastatic Setting. Biomolecules 2023; 13:689. [PMID: 37189436 PMCID: PMC10136331 DOI: 10.3390/biom13040689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
With the development of immune checkpoint inhibitors (ICIs), the tumour immune microenvironment (TIME) has been increasingly considered to improve cancer management. The TIME of metastatic lesions is strongly influenced by the underlying immune contexture of the organ in which they are located. The metastatic location itself appears to be an important prognostic factor in predicting outcomes after ICI treatment in cancer patients. Patients with liver metastases are less likely to respond to ICIs than patients with metastases in other organs, likely due to variations in the metastatic TIME. Combining additional treatment modalities is an option to overcome this resistance. Radiotherapy (RT) and ICIs have been investigated together as an option to treat various metastatic cancers. RT can induce a local and systemic immune reaction, which can promote the patient's response to ICIs. Here, we review the differential impact of the TIME according to metastatic location. We also explore how RT-induced TIME modifications could be modulated to improve outcomes of RT-ICI combinations.
Collapse
Affiliation(s)
- Julien Pierrard
- UCLouvain, Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institute de Recherche Experimentale et Clinique (IREC), 1200 Brussels, Belgium
- Radiation Oncology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Geneviève Van Ooteghem
- UCLouvain, Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institute de Recherche Experimentale et Clinique (IREC), 1200 Brussels, Belgium
- Radiation Oncology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Marc Van den Eynde
- UCLouvain, Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institute de Recherche Experimentale et Clinique (IREC), 1200 Brussels, Belgium
- Medical Oncology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
31
|
Real Check RIO: A Real-World Analysis of Nivolumab in First Line Metastatic Melanoma Assessing Efficacy, Safety and Predictive Factors. Cancers (Basel) 2023; 15:cancers15041265. [PMID: 36831607 PMCID: PMC9953812 DOI: 10.3390/cancers15041265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
We performed a retrospective study on 51 metastatic melanoma patients treated with Nivolumab in first line, at the Regional Institute of Oncology (RIO) Iasi, Romania between April 2017 and December 2019. We studied the efficacy and safety of anti-PD-1 immune checkpoint inhibitor therapy on a treatment-naive population. After a median follow-up of 36 months, the median progression free survival (PFS) was 26 months (95% CI, 15-36) and the median overall survival (OS) was 31 months (95% CI, 20.1-41.8). At 12 months after the initiation of immunotherapy, the percentage of patients alive was 70%, and at 24 months 62.5%. The most common adverse events observed were dermatological (23.5%) and grade ≥3 was identified in 4 (6.8%) patients. Multivariate analysis indicated that the presence of liver metastases (HR 4.42; 95% CI: 1.88-10.4, p = 0.001) and a neutrophils/lymphocytes ratio (NLR) were associated with poor survival (HR 3.21; 95% CI: 1.04-9.87, p = 0.04). Although retrospective data on a small group of patients were analyzed, we can conclude that our results in RIO are similar to those described in clinical trials and other real-world studies. Our study highlights the potential usefulness of liver metastases and NLR as novel predictive factors in clinical decision-making.
Collapse
|
32
|
Ben-David R, Veredgorn Y, Savin Z, Bar-Yosef Y, Yossepowitch O, Sofer M, Mano R. External validation of a simplified prognostic model for survival in patients with extrinsic malignant ureteral obstruction treated with tandem ureteral stents - a retrospective cohort study. Scand J Urol 2023; 57:90-96. [PMID: 36708159 DOI: 10.1080/21681805.2023.2171113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Prognostic models of survival can identify patients with extrinsic malignant ureteral obstruction who will benefit from long-term drainage as offered by tandem ureteral stents. The study aims to validate a simplified prognostic model published by Cordeiro et al. and to identify additional prognostic predictors in a cohort of patients drained solely with tandem ureteral stents. METHODS Medical records of consecutive patients who underwent drainage of malignant ureteral obstruction with tandem ureteral stents between 2007 and 2020 were reviewed retrospectively; patients with benign ureteral obstruction were excluded. Risk factors for survival included were: [1] the number of malignancy-related events (categorized as ≥4 and <4) and [2] the Eastern Cooperative Oncology Group Index (categorized as ≥2 and <2)]. Patients with ≥1 risk factor were grouped as intermediate-unfavorable risk and those without risk factors as favorable risk. The Kaplan-Meier and log-rank tests were used for survival analysis. Univariable and multivariable Cox regression analyses were used to identify predictors of outcome. RESULTS The study cohort consisted of 65 patients; the median age was 60 years (IQR 51-72). The median follow-up time from diagnosis of hydronephrosis was 51 months (IQR 38-64). Estimated probabilities of survival at 1 month, 6 months 1 year, and 2 years were 100%, 87%, 75% and 57%, respectively in the favorable risk group (n = 40), and in the intermediate-unfavorable risk group (n = 25), 96%, 72%, 52%, and 20%, respectively, (p = .003). On multivariable analysis, the presence of ≥4 malignancy-related events (HR = 2.04, 95% CI [1.07-3.86], p = .03) and lung metastasis (HR = 2.37, 95% CI [1.0-5.6], p = .05) were associated with shorter survival. CONCLUSIONS Our findings validate the prognostic model published by Cordeiro et al. The model can be applied when counseling patients being considered for drainage with tandem ureteral stents.
Collapse
Affiliation(s)
- Reuben Ben-David
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yotam Veredgorn
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ziv Savin
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Bar-Yosef
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Urology, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ofer Yossepowitch
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mario Sofer
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roy Mano
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Gata6 + large peritoneal macrophages: an evolutionarily conserved sentinel and effector system for infection and injury. Trends Immunol 2023; 44:129-145. [PMID: 36623953 DOI: 10.1016/j.it.2022.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 01/08/2023]
Abstract
There are striking similarities between the sea urchin cavity macrophage-like phagocytes (coelomocytes) and mammalian cavity macrophages in not only their location, but also their behaviors. These cells are crucial for maintaining homeostasis within the cavity following a breach, filling the gap and functioning as a barrier between vital organs and the environment. In this review, we summarize the evolving literature regarding these Gata6+ large peritoneal macrophages (GLPMs), focusing on ontogeny, their responses to perturbations, including their rapid aggregation via coagulation, as well as scavenger receptor cysteine-rich domains and their potential roles in diseases, such as cancer. We challenge the 50-year old phenomenon of the 'macrophage disappearance reaction' (MDR) and propose the new term 'macrophage disturbance of homeostasis reaction' (MDHR), which may better describe this complex phenomenon.
Collapse
|
34
|
Beypınar I, Sözel Y, Önder AH. Assessing the prognostic value of IMDC risk score for nivolumab-treated patients with renal cancer and malignant melanoma. Cancer Biomark 2023; 38:367-377. [PMID: 37718781 DOI: 10.3233/cbm-230159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
BACKGROUND The response of Renal Cell Cancer (RCC) to tyrosine kinase inhibitors (TKI) has been well established. Although these stratifications have been established for TKI response and prognosis, these parameters have recently been used to predict immunotherapy response in RCC. We aimed to use a combination of clinical parameters of International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk groups and metastatic sites at the time of diagnosis to predict the effectiveness of immune checkpoint inhibitors in malignant melanoma (MM). METHOD In this cross-sectional study, we retrospectively analyzed the demographic information, metastatic sites, and IMDC risk group data. The blood parameters were included in the first cycle of nivolumab treatment. RESULTS The OS was statistically different between the RCC and MM groups in terms of the IMDC. In univariate analysis of stage at diagnosis, CRP levels and bone and bone marrow metastases were confirmed to be prognostic factors in the MM population in terms of OS. Brain metastasis was a prognostic factor for RCC, whereas sex, line of treatment, LDH, bone, and splenic metastasis remained significant in patients with MM in terms of OS. Brain metastasis was prognostic in both cancer types in multivariate analysis in terms of PFS. In addition to brain metastasis, LDH levels and lung, liver, and splenic metastases also affect PFS in patients with MM undergoing nivolumab treatment. CONCLUSION In our study, the IMDC was confirmed to be a prognostic factor for MM. The IMDC groups were similar, except for the favorable RCC and MM groups. Different metastatic sites were prognostic, similar to the IMDC risk group in the MM group.
Collapse
Affiliation(s)
- Ismail Beypınar
- Department of Medical Oncology, Alanya Alaaddin Keykubat University, Alanya/Antalya, Turkey
| | - Yıldız Sözel
- Department of Radiology, Antalya Research and Education Hospital, Antalya, Turkey
| | - Arif Hakan Önder
- Department of Medical Oncology, Antalya Research and Education Hospital, Antalya, Turkey
| |
Collapse
|
35
|
Da L, Qu Z, Zhang C, Shen Y, Huang W, Zhang Y, Gu K. Prognostic value of inflammatory markers and clinical features for survival in advanced or metastatic esophageal squamous cell carcinoma patients receiving anti-programmed death 1 treatment. Front Oncol 2023; 13:1144875. [PMID: 37035159 PMCID: PMC10076857 DOI: 10.3389/fonc.2023.1144875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Purpose This study aims to assess the prognostic value of inflammatory markers and clinical features in advanced or metastatic esophageal squamous cell carcinoma (ESCC) patients receiving anti-programmed death 1 (PD-1) treatment. Methods Based on receiver operating characteristic curve (ROC) analysis, Youden's indexes were applied to determine the cut-off values for inflammatory markers, including neutrophil-to-lymphocyte ratio (NLR), derived neutrophil-to-lymphocye ratio (dNLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII). Wilcoxon test was conducted to evaluate the changes in above inflammatory markers. Kaplan-Meier method was utilized to estimate progression-free survival (PFS) and overall survival (OS), and the Log-rank test was used to compare the different survival between groups. Univariate and multivariate Cox regression analyses were performed to assess the prognostic value of inflammatory markers and clinical features. Results 162 advanced or metastatic ESCC patients receiving anti-PD-1 treatment were enrolled in this retrospective study. The cut-off values of NLR, dNLR, MLR, PLR, and SII were 4.748, 2.214, 0.309, 250.505, and 887.895, respectively. NLR, dNLR, PLR, and SII declined significantly among the partial response (PR) (P<0.001, P<0.001, P=0.036, P<0.001), objective response rate (ORR) (P<0.001, P<0.001, P=0.036, P<0.001), and disease control rate (DCR) (P<0.001, P<0.001, P=0.038, P<0.001) groups, respectively. Significant increases were found in NLR (P<0.001), dNLR (P<0.001), MLR (P=0.001), and SII (P=0.024) when anti-PD-1 treatment failed. Multivariate Cox regression analysis indicated that NLR (P<0.001, P=0.002), lymph node metastasis (P=0.013, P=0.001), Eastern Cooperative Oncology Group Performance Status (ECOG PS) (P=0.008, P=0.002), and treatment lines (P=0.037, P=0.048) were significant prognostic indicators of PFS and OS. Additionally, SII (P=0.016) was also significantly related to OS in ESCC patients. The risk score model showed that low risk patients prolonged PFS and OS than those with middle or high risk (P<0.001, P<0.001). Conclusion Inflammatory markers can reflect short-term outcomes of anti-PD-1 treatment for ESCC patients. NLR, lymph node metastases, ECOG PS, and treatment lines are significant prognostic indicators for PFS and OS. And the risk score model constructed based on the above factors has favourable prognostic predictive value.
Collapse
Affiliation(s)
| | | | | | | | | | - Yiyin Zhang
- *Correspondence: Yiyin Zhang, ; Kangsheng Gu,
| | | |
Collapse
|
36
|
Impact of Liver Metastases and Number of Metastatic Sites on Immune-Checkpoint Inhibitors Efficacy in Patients with Different Solid Tumors: A Retrospective Study. Biomedicines 2022; 11:biomedicines11010083. [PMID: 36672591 PMCID: PMC9855949 DOI: 10.3390/biomedicines11010083] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/28/2022] [Accepted: 11/29/2022] [Indexed: 12/31/2022] Open
Abstract
Background: ICIs have dramatically improved patient outcomes in different malignancies. However, the impact of liver metastases (LM) and number of metastatic sites (MS) remains unclear in patients treated with single-agent anti-PD(L)1. Methods: We aimed to assess the prognostic impact of LM and MS number on progression-free survival (PFS) and overall survival (OS) in a large single-arm retrospective multicentric cohort (IMMUCARE) of patients treated with anti-PD(L)-1 for different solid tumors. Results: A total of 759 patients were enrolled from January 2012 to October 2018. The primary tumor types were non-small cell lung cancer (71%), melanoma (19%), or urologic cancer (10%). At the time of ICI initiation, 167 patients (22%) had LM and 370 patients (49%) had more than MS. LM was associated with a shorter median PFS of 1.9 months (95% CI: 1.8−2.5) vs. 4.0 months (95% CI: 3.6−5.4) in patients without LM (p < 0.001). The median OS of patients with LM was of 5.2 months (95% CI: 4.0−7.7) compared with 12.8 months (95% CI: 11.2−15.1) (p < 0.001). Interestingly, LM were not associated with shorter PFS, or OS compared to other MS types (brain, bone, or lung) in patients with only one MS. Patients with multiple MS also had poor clinical outcomes compared to patients with only one MS. The presence of LM and MS number were independent prognostic factors on overall survival. Conclusion: The presence of LM or multiple MS were associated with poorer survival outcomes in patients treated with anti-PD(L)-1.
Collapse
|
37
|
Yin M, Guan S, Ding X, Zhuang R, Sun Z, Wang T, Zheng J, Li L, Gao X, Wei H, Ma J, Huang Q, Xiao J, Mo W. Construction and validation of a novel web-based nomogram for patients with lung cancer with bone metastasis: A real-world analysis based on the SEER database. Front Oncol 2022; 12:1075217. [PMID: 36568214 PMCID: PMC9780685 DOI: 10.3389/fonc.2022.1075217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Purpose Patients with lung cancer with bone metastasis (LCBM) often have a very poor prognosis. The purpose of this study is to characterize the prevalence and associated factors and to develop a prognostic nomogram to predict the overall survival (OS) and cancer-specific survival (CSS) for patients with LCBM using multicenter population-based data. Methods Patients with LCBM at the time of diagnosis were identified using the Surveillance, Epidemiology, and End Results (SEER) Program database of the National Cancer Institute (NCI) from 2010 to 2015. Multivariable and univariate logistic regression analyses were performed to identify factors associated with all-cause mortality and lung cancer (LC)-specific mortality. The performance of the nomograms was evaluated with the calibration curves, area under the curve (AUC), and decision curve analysis (DCA). Kaplan-Meier analysis and log-rank tests were used to estimate the survival times of patients with LCBM. Results We finally identified 26,367 patients with LCBM who were selected for survival analysis. Multivariate analysis demonstrated age, sex, T stage, N stage, grade, histology, radiation therapy, chemotherapy, primary site, primary surgery, liver metastasis, and brain metastasis as independent predictors for LCBM. The AUC values of the nomogram for the OS prediction were 0.755, 0.746, and 0.775 in the training cohort; 0.757, 0.763, and 0.765 in the internal validation cohort; and 0.769, 0.781, and 0.867 in the external validation cohort. For CSS, the values were 0.753, 0.753, and 0.757 in the training cohort; 0.753, 0.753, and 0.757 in the internal validation cohort; and 0.767, 0.774, and 0.872 in the external validation cohort. Conclusions Our study constructs a new prognostic model and clearly presents the clinicopathological features and survival analysis of patients with LCBM. The result indicated that the nomograms had favorable discrimination, good consistency, and clinical benefits in patients. In addition, our constructed nomogram prediction models may assist physicians in evaluating individualized prognosis and deciding on treatment for patients.
Collapse
Affiliation(s)
- Mengchen Yin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Sisi Guan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xing Ding
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ruoyu Zhuang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengwang Sun
- Department of Musculoskeletal Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Tao Wang
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Anhui, China
| | - Jiale Zheng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Li
- Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xin Gao
- Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Haifeng Wei
- Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Junming Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Quan Huang
- Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianru Xiao
- Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China,*Correspondence: Jianru Xiao, ; Wen Mo,
| | - Wen Mo
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Jianru Xiao, ; Wen Mo,
| |
Collapse
|
38
|
Prognostic Hematologic Biomarkers Following Immune Checkpoint Inhibition in Metastatic Uveal Melanoma. Cancers (Basel) 2022; 14:cancers14235789. [PMID: 36497270 PMCID: PMC9738244 DOI: 10.3390/cancers14235789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Background: There is no standardized treatment for metastatic uveal melanoma (MUM) but immune checkpoint inhibitors (ICI) are increasingly used. While ICI has transformed the survival of metastatic cutaneous melanoma, MUM patients do not equally benefit. Factors known to affect ICI response include the hematologic markers, lactate dehydrogenase (LDH) and neutrophil:lymphocyte ratio (NLR). We evaluated the prognostic value of LDH and NLR at the start of ICI and on treatment in MUM. Methods: MUM patients were treated between August 2006 and May 2022 with combination ipilimumab/nivolumab or ipilimumab/nivolumab/pembrolizumab single-agent therapy. Univariable (UVA) and multivariable (MVA) analyses were used to assess the prognostic value of predefined baseline factors on progression-free (PFS) and overall survival (OS). Results: In forty-six patients with MUM treated with ICI, elevated baseline and on-treatment LDH was prognostic for OS (start of ICI, HR (95% CI): 3.6 (1.9−7.0), p < 0.01; on-treatment, HR (95% CI): 3.7 (1.6−8.8), p < 0.01) and PFS (start of ICI, (HR (95% CI): 2.8 (1.5−5.4), p < 0.0001); on-treatment LDH (HR (95% CI): 2.2 (1.1−4.3), p < 0.01). On-treatment NLR was prognostic for PFS (HR (95% CI): 1.9 (1.0−3.9), p < 0.01). On-treatment LDH remained an important contributor to survival on MVA (OS: HR (95% CI): 1.001 (1.00−1.002), p < 0.05); PFS: HR (95% CI): 1.001 (1.00−1.002), p < 0.01). Conclusions: This study demonstrates that LDH and NLR could be useful in the prognostication of MUM patients treated with ICI. Additional studies are needed to confirm the importance of these and other prognostic biomarkers.
Collapse
|
39
|
Nie C, Lv H, Chen B, Xu W, Wang J, Liu Y, Wang S, Zhao J, He Y, Chen X. Microsatellite stable metastatic colorectal cancer without liver metastasis may be preferred population for regorafenib or fruquintinib plus sintilimab as third-line or above therapy:A real-world study. Front Oncol 2022; 12:917353. [PMID: 36226061 PMCID: PMC9549285 DOI: 10.3389/fonc.2022.917353] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives The antitumor activity of nivolumab plus regorafenib in colorectal cancer from a phase Ib REGONIVO study is encouraging. The present study was conducted to evaluate the efficacy and safety of regorafenib or fruquintinib plus sintilimab as third-line or above therapy in patients with microsatellite stable (MSS) metastatic colorectal cancer. Methods Patients with MSS metastatic colorectal cancer who have failed from prior treatment and received regorafenib or fruquintinib plus sintilimab as third-line or above therapy from January 2019 to December 2020 were prospectively analyzed based on real-world clinical practice. The primary end point was progression free survival (PFS). Secondary end points included objective response rate (ORR), disease control rate (DCR), overall survival (OS), and safety. Results 42 patients received regorafenib plus sintilimab(RS), and the other 30 patients received fruquintinib plus sintilimab(FS). In the general population, the ORR and DCR were 13.9% and 70.8%, and the median PFS and OS was 4.2(95% CI=2.9-5.5) and 10.5 (95% CI=8.6-12.4) months, respectively. There were no statistically significant differences between RS and FS group in PFS (3.5(2.2-4.8) vs. 5.5(3.5-7.5) months, P=0.434) and OS (11.0(7.0-15.0) vs. 10.5(3.8-17.2) months, P=0.486). Subgroup analysis suggested that patients without liver metastasis responded well to this combination regimen (ORR: 21.4% vs. 9.1%) and obtained better OS (26(8.8-43.2) vs. 10.0(7.4-12.6) months, P=0.016). The incidence of Grade 3-4 adverse events (AEs) was 15.3% and the toxicities were generally tolerable and manageable. Conclusions Regorafenib or fruquintinib plus sintilimab as third-line or above therapy provide a feasible treatment regimen for MSS metastatic colorectal cancer with tolerated toxicity. Patients without liver metastasis may be the preferred population for this combination regimen.
Collapse
Affiliation(s)
- Caiyun Nie
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Huifang Lv
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Weifeng Xu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Jianzheng Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Yingjun Liu
- Department of General Surgery, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Saiqi Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Jing Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Yunduan He
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, China
- *Correspondence: Xiaobing Chen,
| |
Collapse
|
40
|
Ter Maat LS, van Duin IAJ, Elias SG, van Diest PJ, Pluim JPW, Verhoeff JJC, de Jong PA, Leiner T, Veta M, Suijkerbuijk KPM. Imaging to predict checkpoint inhibitor outcomes in cancer. A systematic review. Eur J Cancer 2022; 175:60-76. [PMID: 36096039 DOI: 10.1016/j.ejca.2022.07.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Checkpoint inhibition has radically improved the perspective for patients with metastatic cancer, but predicting who will not respond with high certainty remains difficult. Imaging-derived biomarkers may be able to provide additional insights into the heterogeneity in tumour response between patients. In this systematic review, we aimed to summarise and qualitatively assess the current evidence on imaging biomarkers that predict response and survival in patients treated with checkpoint inhibitors in all cancer types. METHODS PubMed and Embase were searched from database inception to 29th November 2021. Articles eligible for inclusion described baseline imaging predictive factors, radiomics and/or imaging machine learning models for predicting response and survival in patients with any kind of malignancy treated with checkpoint inhibitors. Risk of bias was assessed using the QUIPS and PROBAST tools and data was extracted. RESULTS In total, 119 studies including 15,580 patients were selected. Of these studies, 73 investigated simple imaging factors. 45 studies investigated radiomic features or deep learning models. Predictors of worse survival were (i) higher tumour burden, (ii) presence of liver metastases, (iii) less subcutaneous adipose tissue, (iv) less dense muscle and (v) presence of symptomatic brain metastases. Hazard rate ratios did not exceed 2.00 for any predictor in the larger and higher quality studies. The added value of baseline fluorodeoxyglucose positron emission tomography parameters in predicting response to treatment was limited. Pilot studies of radioactive drug tracer imaging showed promising results. Reports on radiomics were almost unanimously positive, but numerous methodological concerns exist. CONCLUSIONS There is well-supported evidence for several imaging biomarkers that can be used in clinical decision making. Further research, however, is needed into biomarkers that can more accurately identify which patients who will not benefit from checkpoint inhibition. Radiomics and radioactive drug labelling appear to be promising approaches for this purpose.
Collapse
Affiliation(s)
- Laurens S Ter Maat
- Image Science Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Isabella A J van Duin
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Josien P W Pluim
- Image Science Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Medical Image Analysis, Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Joost J C Verhoeff
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Pim A de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Tim Leiner
- Utrecht University, Utrecht, the Netherlands; Department of Radiology, Mayo Clinical, Rochester, MN, USA
| | - Mitko Veta
- Medical Image Analysis, Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
41
|
Küstner T, Vogel J, Hepp T, Forschner A, Pfannenberg C, Schmidt H, Schwenzer NF, Nikolaou K, la Fougère C, Seith F. Development of a Hybrid-Imaging-Based Prognostic Index for Metastasized-Melanoma Patients in Whole-Body 18F-FDG PET/CT and PET/MRI Data. Diagnostics (Basel) 2022; 12:2102. [PMID: 36140504 PMCID: PMC9498091 DOI: 10.3390/diagnostics12092102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Besides tremendous treatment success in advanced melanoma patients, the rapid development of oncologic treatment options comes with increasingly high costs and can cause severe life-threatening side effects. For this purpose, predictive baseline biomarkers are becoming increasingly important for risk stratification and personalized treatment planning. Thus, the aim of this pilot study was the development of a prognostic tool for the risk stratification of the treatment response and mortality based on PET/MRI and PET/CT, including a convolutional neural network (CNN) for metastasized-melanoma patients before systemic-treatment initiation. The evaluation was based on 37 patients (19 f, 62 ± 13 y/o) with unresectable metastasized melanomas who underwent whole-body 18F-FDG PET/MRI and PET/CT scans on the same day before the initiation of therapy with checkpoint inhibitors and/or BRAF/MEK inhibitors. The overall survival (OS), therapy response, metastatically involved organs, number of lesions, total lesion glycolysis, total metabolic tumor volume (TMTV), peak standardized uptake value (SULpeak), diameter (Dmlesion) and mean apparent diffusion coefficient (ADCmean) were assessed. For each marker, a Kaplan−Meier analysis and the statistical significance (Wilcoxon test, paired t-test and Bonferroni correction) were assessed. Patients were divided into high- and low-risk groups depending on the OS and treatment response. The CNN segmentation and prediction utilized multimodality imaging data for a complementary in-depth risk analysis per patient. The following parameters correlated with longer OS: a TMTV < 50 mL; no metastases in the brain, bone, liver, spleen or pleura; ≤4 affected organ regions; no metastases; a Dmlesion > 37 mm or SULpeak < 1.3; a range of the ADCmean < 600 mm2/s. However, none of the parameters correlated significantly with the stratification of the patients into the high- or low-risk groups. For the CNN, the sensitivity, specificity, PPV and accuracy were 92%, 96%, 92% and 95%, respectively. Imaging biomarkers such as the metastatic involvement of specific organs, a high tumor burden, the presence of at least one large lesion or a high range of intermetastatic diffusivity were negative predictors for the OS, but the identification of high-risk patients was not feasible with the handcrafted parameters. In contrast, the proposed CNN supplied risk stratification with high specificity and sensitivity.
Collapse
Affiliation(s)
- Thomas Küstner
- MIDAS.Lab, Department of Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| | - Jonas Vogel
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University Hospital Tübingen, 72076 Tubingen, Germany
| | - Tobias Hepp
- MIDAS.Lab, Department of Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital of Tübingen, 72070 Tubingen, Germany
| | - Christina Pfannenberg
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| | - Holger Schmidt
- Faculty of Medicine, Eberhard-Karls-University Tübingen, 72076 Tubingen, Germany
- Siemens Healthineers, 91052 Erlangen, Germany
| | - Nina F. Schwenzer
- Faculty of Medicine, Eberhard-Karls-University Tübingen, 72076 Tubingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies, Eberhard Karls University, 72076 Tubingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, 72076 Tubingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University Hospital Tübingen, 72076 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies, Eberhard Karls University, 72076 Tubingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, 72076 Tubingen, Germany
| | - Ferdinand Seith
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| |
Collapse
|
42
|
Qin Q, Jun T, Wang B, Patel VG, Mellgard G, Zhong X, Gogerly-Moragoda M, Parikh AB, Leiter A, Gallagher EJ, Alerasool P, Garcia P, Joshi H, Galsky M, Oh WK, Tsao CK. Clinical factors associated with outcome in solid tumor patients treated with immune-checkpoint inhibitors: a single institution retrospective analysis. Discov Oncol 2022; 13:73. [PMID: 35960456 PMCID: PMC9374856 DOI: 10.1007/s12672-022-00538-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/02/2022] [Indexed: 04/17/2023] Open
Abstract
OBJECTIVES Response to immune checkpoint inhibitor (ICI) remains limited to a subset of patients and predictive biomarkers of response remains an unmet need, limiting our ability to provide precision medicine. Using real-world data, we aimed to identify potential clinical prognosticators of ICI response in solid tumor patients. METHODS We conducted a retrospective analysis of all solid tumor patients treated with ICIs at the Mount Sinai Hospital between January 2011 and April 2017. Predictors assessed included demographics, performance status, co-morbidities, family history of cancer, smoking status, cancer type, metastatic pattern, and type of ICI. Outcomes evaluated include progression free survival (PFS), overall survival (OS), overall response rate (ORR) and disease control rate (DCR). Univariable and multivariable Cox proportional hazard models were constructed to test the association of predictors with outcomes. RESULTS We identified 297 ICI-treated patients with diagnosis of non-small cell lung cancer (N = 81, 27.3%), melanoma (N = 73, 24.6%), hepatocellular carcinoma (N = 51, 17.2%), urothelial carcinoma (N = 51, 17.2%), head and neck squamous cell carcinoma (N = 23, 7.7%), and renal cell carcinoma (N = 18, 6.1%). In multivariable analysis, good performance status of ECOG ≤ 2 (PFS, ORR, DCR and OS) and family history of cancer (ORR and DCR) associated with improved ICI response. Bone metastasis was associated with worse outcomes (PFS, ORR, and DCR). CONCLUSIONS Mechanisms underlying the clinical predictors of response observed in this real-world analysis, such as genetic variants and bone metastasis-tumor microenvironment, warrant further exploration in larger studies incorporating translational endpoints. Consistently positive clinical correlates may help inform patient stratification when considering ICI therapy.
Collapse
Affiliation(s)
- Qian Qin
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | - Tomi Jun
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | - Bo Wang
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | - Vaibhav G Patel
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | | | - Xiaobo Zhong
- Department of Population Health and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anish B Parikh
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center-James Cancer Hospital, Columbus, OH, USA
| | - Amanda Leiter
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily J Gallagher
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Parissa Alerasool
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
- New York Medical College, Valhalla, NY, USA
| | - Philip Garcia
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | | | - Matthew Galsky
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | - William K Oh
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA
| | - Che-Kai Tsao
- Division of Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Third Floor Admin Suite #108 (Hess Building), New York, NY, USA.
| |
Collapse
|
43
|
Gata6 + resident peritoneal macrophages promote the growth of liver metastasis. Nat Commun 2022; 13:4406. [PMID: 35906202 PMCID: PMC9338095 DOI: 10.1038/s41467-022-32080-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 07/18/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence suggests that resident macrophages within tissues are enablers of tumor growth. However, a second population of resident macrophages surrounds all visceral organs within the cavities and nothing is known about these GATA6+ large peritoneal macrophages (GLPMs) despite their ability to invade injured visceral organs by sensing danger signals. Here, we show that GLPMs invade growing metastases that breach the visceral mesothelium of the liver via the "find me signal", ATP. Depleting GLPMs either by pharmacological or genetic tools, reduces metastases growth. Apoptotic bodies from tumor cells induces programmed cell death ligand 1 (PD-L1) upregulation on GLPMs which block CD8+ T cell function. Direct targeting of GLPMs by intraperitoneal but not intravenous administration of anti-PD-L1 reduces tumor growth. Thermal ablation of liver metastases recruits huge numbers of GLPMs and enables rapid regrowth of tumors. GLPMs contribute to metastatic growth and tumor recurrence.
Collapse
|
44
|
Organ-specific efficacy in advanced non-small cell lung cancer patients treated with first-line single-agent immune checkpoint inhibitors. Chin Med J (Engl) 2022; 135:1404-1413. [PMID: 35869859 PMCID: PMC9481449 DOI: 10.1097/cm9.0000000000002217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: Methods: Results: Conclusions:
Collapse
|
45
|
Kim HS, Kim CG, Hong JY, Kim IH, Kang B, Jung S, Kim C, Shin SJ, Choi HJ, Cheon J, Chon HJ, Lim HY. The presence and size of intrahepatic tumors determine the therapeutic efficacy of nivolumab in advanced hepatocellular carcinoma. Ther Adv Med Oncol 2022; 14:17588359221113266. [PMID: 35860833 PMCID: PMC9290164 DOI: 10.1177/17588359221113266] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose: Inter-tumoral heterogeneity at the differential lesion level raises the possibility of distinct organ-specific responses to immune checkpoint inhibitors (ICIs). We aimed to comprehensively examine the clinicopathological factors to predict and assess the efficacy of nivolumab, programmed cell death protein 1 (PD-1) blockade at an individual tumor site-specific level in patients with advanced hepatocellular carcinoma (aHCC). Patients and Methods: We enrolled 261 aHCC patients treated with nivolumab between 2012 and 2018. Eighty-one clinicopathological factors were comprehensively collected and analyzed. The association between all variables and survival outcomes was evaluated. According to tumor site, the organ-specific responses were assessed based on the Response Evaluation Criteria in Solid Tumors, version 1.1. Results: The liver was the most commonly involved organ (75.1%), followed by the lungs (37.5%) and lymph nodes (LNs, 11.5%). The liver of nonresponders was more frequently the organ of progression, while the lungs of responders were more frequently the organs of response. Among the 455 individual lesions (liver, n = 248; lung, n = 124; LN, n = 35; others including bone or soft tissues, n = 48), intrahepatic tumors showed the least response (10.1%), followed by lung (24.2%) and LN tumors (37.1%), indicating the presence of distinct organ-specific responses to nivolumab. In intrahepatic tumors, the organ-specific response rate decreased as the size increased (13% for ⩽50 mm, 8.1% for 50–100 mm, and 5.5% for >100 mm). In the subgroup analysis according to tumor location, patients with lung only metastasis (⩾30 mm) showed the best progression-free survival (PFS) and overall survival (OS). In contrast, primary HCC (⩾100 mm) without lung metastasis had the worst PFS and OS. Comprehensive analyses also revealed that liver function and systemic inflammatory indices, such as neutrophil-to-lymphocyte ratio (NLR), were significantly associated with PFS and OS. Conclusion: The presence and size of liver tumors, liver function, and NLR are key factors determining the response to nivolumab in aHCC. These clinical factors should be considered when treating patients with advanced HCC with PD-1 blockade.
Collapse
Affiliation(s)
- Han Sang Kim
- Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Gon Kim
- Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Il-Hwan Kim
- Department of Oncology, Haeundae Paik Hospital, Cancer Center, Inje University College of Medicine, Busan, Korea
| | - Beodeul Kang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea
| | - Sanghoon Jung
- Department of Radiology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea
| | - Sang Joon Shin
- Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jin Choi
- Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jaekyung Cheon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Gyeonggi-do, Korea
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Gyeonggi-do, Korea
| | - Ho Yeong Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06171, Republic of Korea
| |
Collapse
|
46
|
Zhang T, Jia Y, Yu Y, Zhang B, Xu F, Guo H. Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev 2022; 186:114319. [PMID: 35545136 DOI: 10.1016/j.addr.2022.114319] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Immunotherapy based on immune checkpoint inhibitors has evolved into a new pillar of cancer treatment in clinics, but dealing with treatment resistance (either primary or acquired) is a major challenge. The tumor microenvironment (TME) has a substantial impact on the pathological behaviors and treatment response of many cancers. The biophysical clues in TME have recently been considered as important characteristics of cancer. Furthermore, there is mounting evidence that biophysical cues in TME play important roles in each step of the cascade of cancer immunotherapy that synergistically contribute to immunotherapy resistance. In this review, we summarize five main biophysical cues in TME that affect resistance to immunotherapy: extracellular matrix (ECM) structure, ECM stiffness, tumor interstitial fluid pressure (IFP), solid stress, and vascular shear stress. First, the biophysical factors involved in anti-tumor immunity and therapeutic antibody delivery processes are reviewed. Then, the causes of these five biophysical cues and how they contribute to immunotherapy resistance are discussed. Finally, the latest treatment strategies that aim to improve immunotherapy efficacy by targeting these biophysical cues are shared. This review highlights the biophysical cues that lead to immunotherapy resistance, also supplements their importance in related technologies for studying TME biophysical cues in vitro and therapeutic strategies targeting biophysical cues to improve the effects of immunotherapy.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuanbo Jia
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yang Yu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
47
|
Msaouel P, Goswami S, Thall PF, Wang X, Yuan Y, Jonasch E, Gao J, Campbell MT, Shah AY, Corn PG, Tam AL, Ahrar K, Rao P, Sircar K, Cohen L, Basu S, Duan F, Jindal S, Zhang Y, Chen H, Yadav SS, Shazer R, Der-Torossian H, Allison JP, Sharma P, Tannir NM. A phase 1-2 trial of sitravatinib and nivolumab in clear cell renal cell carcinoma following progression on antiangiogenic therapy. Sci Transl Med 2022; 14:eabm6420. [PMID: 35442707 PMCID: PMC11924199 DOI: 10.1126/scitranslmed.abm6420] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The accumulation of immune-suppressive myeloid cells is a critical determinant of resistance to anti-programmed death-1 (PD-1) therapy in advanced clear cell renal cell carcinoma (ccRCC). In preclinical models, the tyrosine kinase inhibitor sitravatinib enhanced responses to anti-PD-1 therapy by modulating immune-suppressive myeloid cells. We conducted a phase 1-2 trial to choose an optimal sitravatinib dose combined with a fixed dose of nivolumab in 42 immunotherapy-naïve patients with ccRCC refractory to prior antiangiogenic therapies. The combination demonstrated no unexpected toxicities and achieved an objective response rate of 35.7% and a median progression-free survival of 11.7 months, with 80.1% of patients alive after a median follow-up of 18.7 months. Baseline peripheral blood neutrophil-to-lymphocyte ratio correlated with response to sitravatinib and nivolumab. Patients with liver metastases showed durable responses comparable to patients without liver metastases. In addition, correlative studies demonstrated reduction of immune-suppressive myeloid cells in the periphery and tumor microenvironment following sitravatinib treatment. This study provides a rationally designed combinatorial strategy to improve outcomes of anti-PD-1 therapy in advanced ccRCC.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sangeeta Goswami
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peter F Thall
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuemei Wang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying Yuan
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amishi Yogesh Shah
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paul Gettys Corn
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alda L Tam
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kamran Ahrar
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Priya Rao
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kanishka Sircar
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lorenzo Cohen
- Department of Palliative, Rehabilitation and Integrative Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sreyashi Basu
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Fei Duan
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sonali Jindal
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuwei Zhang
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Chen
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shalini S Yadav
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | - James P Allison
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
48
|
An A, Hui D. Immunotherapy Versus Hospice: Treatment Decision-Making in the Modern Era of Novel Cancer Therapies. Curr Oncol Rep 2022; 24:285-294. [PMID: 35113356 DOI: 10.1007/s11912-022-01203-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW Modern advances in cancer treatment with immunotherapy have created substantial hope for patients and oncologists alike due to a new possibility of durable response which can verge on "cure." This, in combination with a more favorable toxicity profile, has led many oncologists to consider immunotherapy for patients who might have previously been recommended for hospice. In this narrative review, we discuss (1) the risks and benefits of immunotherapy in patients with far advanced cancer in the last months of life, (2) the role of supportive and palliative care, and (3) how to navigate complex treatment decisions for these patients. RECENT FINDINGS Unfortunately, data on immunotherapy outcomes for patients with poor performance status and far advanced disease are quite limited. Where available, studies consistently report poorer survival outcomes compared to patients with preserved performance status. However, a minority of patients (15-30%) may achieve at least partial response with immunotherapy, which can be quite durable. Such prognostic uncertainty leads to additional challenges in treatment discussions and decision-making. Given such prognostic uncertainty, clinicians should individualize treatment with consideration for all the various factors that may inform each patient's expected outcome with immunotherapy. Early involvement of palliative care in the disease trajectory can help patients with advanced cancer to optimize their quality of life, improve illness understanding, navigate prognostic uncertainty, and facilitate complex decision-making regarding cancer treatments. With upfront, open discussions of immunotherapy expectations, oncologists can help ensure treatments are aligned with patient goals and optimize value outcomes.
Collapse
Affiliation(s)
- Amy An
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 463, Houston, TX, 77030, USA.
| | - David Hui
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
49
|
He K, Barsoumian HB, Bertolet G, Verma V, Leuschner C, Koay EJ, Ludmir EB, Hsu E, Pisipati E, Voss TA, Puebla-Osorio N, Cortez MA, Welsh JW. Novel Use of Low-Dose Radiotherapy to Modulate the Tumor Microenvironment of Liver Metastases. Front Immunol 2022; 12:812210. [PMID: 34975924 PMCID: PMC8714746 DOI: 10.3389/fimmu.2021.812210] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Despite multiple therapeutic approaches, the presence of liver metastases carries a guarded prognosis, urgently necessitating further clinical and scientific research to develop curative interventions. The liver is an immunoprivileged organ that suppresses the effectiveness of immunotherapies in patients with hepatic metastases. Cancer immunotherapies have been successfully bolstered by low-dose radiotherapy (LDRT), which is capable of reprogramming the tumor microenvironment (TME) from an immunosuppressive to an immunostimulatory one. Likewise, LDRT may be able to revoke the immune privilege enjoyed by the liver, permitting successful immunotherapies there. Here, we first review challenges that face the treatment of liver metastases. We next outline emerging preclinical and clinical evidence supporting enhanced systemic tumor control of LDRT in the context of cancer immunotherapy. Finally, we will discuss the rationale of combining liver-directed LDRT with immunostimulatory strategies to overcome immune resistance and achieve better clinical response. This notion is supported by a recent case study in which a patient who had progressed following T cell therapy experienced a complete response after LDRT to the liver.
Collapse
Affiliation(s)
- Kewen He
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Genevieve Bertolet
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Carola Leuschner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan B Ludmir
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan Hsu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Esha Pisipati
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tiffany A Voss
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
50
|
Sagie S, Gadot M, Levartovsky M, Gantz Sorotsky H, Berger R, Sarfaty M, Percik R. Immune-Related Thyroiditis as a Predictor for Survival in Metastatic Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14040875. [PMID: 35205622 PMCID: PMC8870210 DOI: 10.3390/cancers14040875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/30/2022] [Accepted: 02/06/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary In this study, we evaluated the association of immune mediated thyroid dysfunction (irT) with survival in 123 metastatic renal cell carcinoma patients treated with immunotherapy in a single center. We found that irT is a prevalent and early event associated with prolonged survival in high-risk patients. Abstract Immune checkpoint inhibitors (CPI) are indicated for metastatic renal cell carcinoma (mRCC). Immune-related thyroiditis (irT), an immune-related adverse event (irAE), affects up to 30% of patients. We aimed to determine whether irT is associated with overall survival in mRCC. A retrospective cohort study of 123 consecutive patients treated with CPI for mRCC in a single center between 2015 and 2020 was conducted. Disease risk stratification was assessed by two methods: Heng criteria and a novel dichotomic stratification system to “Low risk” versus “High risk” adding number of metastatic sites. Thirty-eight percent of patients developed irT. In the general cohort, irT was not associated with a survival benefit. However, irT was associated with better survival in the poor risk group per Heng criteria (n = 17, HR = 0.25, p = 0.04) and in the novel “High risk” group (HR = 0.28, n = 42, p = 0.01), including after accounting for covariates in multivariate analysis (HR = 0.27, p = 0.003). Having any irAE was associated with improved survival in the whole cohort, with no significant correlation of any specific irAE, in either the whole cohort or the “High risk” group. We conclude that irT is an early and prevalent irAE, associated with prolonged survival in patients with poor/“High” risk mRCC.
Collapse
Affiliation(s)
- Shira Sagie
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sheba Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Moran Gadot
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Meital Levartovsky
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Hadas Gantz Sorotsky
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Raanan Berger
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Michal Sarfaty
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ruth Percik
- Institute of Oncology, Sheba Medical Center, Ramat Gan 52621, Israel; (S.S.); (M.G.); (M.L.); (H.G.S.); (R.B.); (M.S.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan 52621, Israel
- Correspondence: ; Tel.: +972-548118786
| |
Collapse
|