51
|
Masters JC, Khandelwal A, di Pietro A, Dai H, Brar S. Model-informed drug development supporting the approval of the avelumab flat-dose regimen in patients with advanced renal cell carcinoma. CPT Pharmacometrics Syst Pharmacol 2022; 11:458-468. [PMID: 35166465 PMCID: PMC9007597 DOI: 10.1002/psp4.12771] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 11/29/2022] Open
Abstract
Avelumab is an anti–PD‐L1 monoclonal antibody approved as monotherapy for Merkel cell carcinoma (MCC) and urothelial carcinoma (UC), and in combination with axitinib for advanced renal cell carcinoma (aRCC). Although initially approved with weight‐based dosing (10 mg/kg intravenously [IV] every 2 weeks [Q2W]), avelumab was subsequently approved for flat dosing (800 mg IV Q2W) based on population pharmacokinetic (PopPK), exposure‐efficacy, and exposure‐safety modeling in MCC and UC. Here, through modeling and simulation, we provide justification for a flat‐dose regimen of avelumab plus axitinib in aRCC. Simulated exposure metrics from the previous monotherapy PopPK model (1827 patients) for both weight‐based and flat‐dose regimens were compared with exposure metrics from treatment‐naive patients with aRCC who received avelumab plus axitinib (488 patients). The aRCC population exposures were derived from a fit‐for‐purpose PopPK model developed using data from monotherapy and combination studies and the existing base structural PopPK model. Exposure‐response relationships for safety were analyzed, including grade ≥3 treatment‐emergent adverse events (TEAEs), any‐grade infusion‐related reactions, and TEAE any‐grade immune‐related adverse events (irAEs). Weight‐based dosing of avelumab in the aRCC population yielded similar PK exposures to the flat‐dose regimen reference exposures in the monotherapy population. Increased avelumab exposure was not associated with increased probabilities of grade ≥3 TEAEs or any‐grade IRRs, although there was a weak association with an increased probability of any‐grade irAEs. Overall, models in aRCC suggest that the avelumab 800‐mg Q2W flat‐dose regimen would provide similar benefits compared with weight‐based dosing with no meaningful change in the probability of AEs.
Collapse
|
52
|
Sochacka-Ćwikła A, Mączyński M, Regiec A. FDA-Approved Small Molecule Compounds as Drugs for Solid Cancers from Early 2011 to the End of 2021. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072259. [PMID: 35408658 PMCID: PMC9000317 DOI: 10.3390/molecules27072259] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023]
Abstract
Solid cancers are the most common types of cancers diagnosed globally and comprise a large number of deaths each year. The main challenge currently in drug development for tumors raised from solid organs is to find more selective compounds, which exploit specific molecular targets. In this work, the small molecule drugs registered by the Food and Drug Administration (FDA) for solid cancers treatment between 2011 and 2022 were identified and analyzed by investigating a type of therapy they are used for, as well as their structures and mechanisms of action. On average, 4 new small molecule agents were introduced each year, with a few exceptions, for a total of 62 new drug approvals. A total of 50 of all FDA-approved drugs have also been authorized for use in the European Union by the European Medicines Agency (EMA). Our analysis indicates that many more anticancer molecules show a selective mode of action, i.e., 49 targeted agents, 5 hormone therapies and 3 radiopharmaceuticals, compared to less specific cytostatic action, i.e., 5 chemotherapeutic agents. It should be emphasized that new medications are indicated for use mainly for monotherapy and less for a combination or adjuvant therapies. The comprehensive data presented in this review can serve for further design and development of more specific targeted agents in clinical usage for solid tumors.
Collapse
|
53
|
Hu H, Chen Y, Tan S, Wu S, Huang Y, Fu S, Luo F, He J. The Research Progress of Antiangiogenic Therapy, Immune Therapy and Tumor Microenvironment. Front Immunol 2022; 13:802846. [PMID: 35281003 PMCID: PMC8905241 DOI: 10.3389/fimmu.2022.802846] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Anti-angiogenesis therapy, a promising strategy against cancer progression, is limited by drug-resistance, which could be attributed to changes within the tumor microenvironment. Studies have increasingly shown that combining anti-angiogenesis drugs with immunotherapy synergistically inhibits tumor growth and progression. Combination of anti-angiogenesis therapy and immunotherapy are well-established therapeutic options among solid tumors, such as non-small cell lung cancer, hepatic cell carcinoma, and renal cell carcinoma. However, this combination has achieved an unsatisfactory effect among some tumors, such as breast cancer, glioblastoma, and pancreatic ductal adenocarcinoma. Therefore, resistance to anti-angiogenesis agents, as well as a lack of biomarkers, remains a challenge. In this review, the current anti-angiogenesis therapies and corresponding drug-resistance, the relationship between tumor microenvironment and immunotherapy, and the latest progress on the combination of both therapeutic modalities are discussed. The aim of this review is to discuss whether the combination of anti-angiogenesis therapy and immunotherapy can exert synergistic antitumor effects, which can provide a basis to exploring new targets and developing more advanced strategies.
Collapse
Affiliation(s)
- Haoyue Hu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Medicine School of University of Electronic Science and Technology, Chengdu, China
| | - Yue Chen
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Songtao Tan
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Silin Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yan Huang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Shengya Fu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, China
| | - Feng Luo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Jun He
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, China
| |
Collapse
|
54
|
Salloum A, Habre M, Chebl JA, Chebl KA, Atallah C, Medawar G, Kourie HR. Dermatological adverse events associated with immune checkpoint inhibitor-based combinations of anticancer therapies: a systematic review. Immunotherapy 2022; 14:489-503. [PMID: 35232283 DOI: 10.2217/imt-2021-0244] [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: 11/21/2022] Open
Abstract
Aim: This paper presents the reported dermatological adverse events (AEs) associated with approved combinations of immunotherapy with drugs of the same class, or in combination with targeted therapy or chemotherapy. Materials & methods: PubMed was used as an electronic database, and a total of 29 articles were reviewed which reported dermatological AEs following combination therapies with nivolumab, ipilimumab, axitinib, pembrolizumab, lenvatinib, avelumab, atezolizumab, carboplatin, etoposide, paclitaxel, bevacizumab, pemetrexed, cisplatin and durvalumab. Results: The dermatological AEs reported were mutually inclusive and the highest incidence of specific AEs was seen in the following combinations: rash in the nivolumab/ipilimumab and lenvatinib/pembrolizumab combinations, pruritus in the atezolizumab/nab-paclitaxel combination, dry skin and palmar-plantar erythrodysesthesia in the axitinib/pembrolizumab combination, and alopecia and severe skin reactions in the pembrolizumab/carboplatin/paclitaxel combination. Conclusion: Knowledge of such side effects is of benefit when choosing an optimal treatment regimen and should be integrated into the monitoring and follow-up phases of treatment.
Collapse
Affiliation(s)
- Antoine Salloum
- Department of Internal Medicine, Roger Williams Medical Center, RI, USA.,Dermatologic SurgiCenter, Philadelphia, PA, USA
| | - Maya Habre
- Faculty of Medicine, University of Balamand, Beirut, Lebanon.,Department of Dermatology, Saint Georges Hospital University Medical Center, Beirut, Lebanon
| | | | - Karen Abi Chebl
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Carl Atallah
- University of Balamand Faculty of Medicine & Medical Sciences El-Koura, Lebanon
| | - Georgio Medawar
- Department of Internal Medicine, Roger Williams Medical Center, RI, USA
| | - Hampig R Kourie
- Department of Hematology-Oncology, Oncology department, Hotel Dieu de France Hospital, Beirut, Lebanon
| |
Collapse
|
55
|
Guo X, Li S, Tong H, Zhang Y, Ji Y, Zhuang R, Zhang C, You Y, Lu W, Zhou Y. Case Report: Complete Response to Antiangiogenesis and Immune Checkpoint Blockade in an Unresectable MMR-Deficient Leiomyosarcoma Harboring Biallelic Loss of PTEN. Front Oncol 2022; 12:802074. [PMID: 35237514 PMCID: PMC8882578 DOI: 10.3389/fonc.2022.802074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/13/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Leiomyosarcoma (LMS) is a malignant smooth muscle neoplasm, in which the efficacy of immune checkpoint blockade (ICB) is very limited. What is worse, loss of PTEN, known as a negative factor for ICB, frequently occurred in LMS. Seeking new strategies for LMS patients harboring loss of PTEN is important and challenging. CASE PRESENTATION A 42-year-old Chinese male was diagnosed as having unresectable LMS of the iliopsoas. After the failure of two prior chemotherapy regimens, whole-exome sequencing revealed that tumor tissue had high tumor mutation burden (689 Muts), high microsatellite instability, and some somatic mutations, including PTEN (copy number loss and p.N323fs), MSH6 (p.F1088fs), TP53 p.R273C, ASXL1 p.G645fs, ATR p.S1843P, and CDKN2A p.A118P. Then, antiangiogenic agent (pazopanib or anlotinib) plus pembrolizumab was administered from January 2 to August 6, 2018. However, pazopanib was stopped on June 18 due to the grade 2/3 adverse effect of hand-foot skin reaction, and anlotinib was administered. Considering that the tumor shrunk after immunotherapy, he underwent radical resection on September 6, 2018. The final pathological diagnosis confirmed pathologic complete response (CR). Until the latest follow-up (September 15, 2021), no progressive disease was observed and total disease-free survival has exceeded 36 months. CONCLUSION We presented a patient with an unresectable mismatch repair (MMR)-deficient LMS harboring biallelic loss of PTEN who achieved CR from a combination strategy of antiangiogenesis plus pembrolizumab. Such a strategy might be a promising strategy to overcome the ICB resistance caused by the loss of PTEN. Such conclusions need to be further confirmed in further investigations.
Collapse
Affiliation(s)
- Xi Guo
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanxing Tong
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Zhang
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongyuan Zhuang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenlu Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang You
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiqi Lu
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Clinic, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
56
|
Chen BH, Kao CC, Xu T, Yang YN, Cha TL, Tsai YT, Liu SY, Wu ST, Meng E, Tsao CW, Chen CL, Sun GH, Yu DS, Chang SY, Yang MH. Determining programmed cell death ligand 1 expression in circulating tumor cells of patients with clear cell renal cell carcinoma and its correlation with response to programmed cell death protein 1 inhibitors. Int J Urol 2022; 29:947-954. [PMID: 35132699 DOI: 10.1111/iju.14812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/21/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE There is a great interest in determining whether the expression of the programmed cell death ligand 1 is correlated with the efficacy of immune checkpoint inhibitors in patients with clear cell renal cell carcinoma; however, primary tumor biopsies can only provide limited information. Therefore, we explored the expression of programmed cell death ligand 1 on circulating tumor cells, which is a potential predictor of therapeutic response. METHODS Circulating tumor cells were isolated from 20 clear cell renal cell carcinoma patients based on cell surface markers targeting clear cell renal cell carcinoma using IsoFlux device, followed by identification according to cell morphology and immunofluorescence studies. Programmed cell death ligand 1 expression status and clinical correlations were also analyzed. RESULTS Before treatment with programmed cell death protein 1 inhibitors, circulating tumor cells were detected in all patients, ranging from 1 to 22 (median 7), with 75% (15/20) of the patients having programmed cell death ligand 1 + circulating tumor cells. Circulating tumor cell programmed cell death ligand 1 expression did not correlate with the immunohistochemical staining of programmed cell death ligand 1 in primary tumors. During treatment with programmed cell death protein 1 inhibitors, the disease control rate was much higher in the patients harboring programmed cell death ligand 1 + circulating tumor cells (73%, 11/15) than others (20%, 1/5). We also found that changes in total circulating tumor cell numbers and programmed cell death ligand 1 + circulating tumor cell counts correlated well with the disease outcome. CONCLUSION We showed that the presence of programmed cell death ligand 1 + circulating tumor cells before programmed cell death protein 1 inhibition treatment could be a prognosis predictive factor and that the dynamic changes in circulating tumor cell numbers may be used to monitor the therapeutic response. Our study confirms the possibility of programmed cell death ligand 1 + circulating tumor cell detection in clear cell renal cell carcinoma patients' blood samples, which can potentially be used as an individualized immunotherapy molecular biomarker for real-time exploration.
Collapse
Affiliation(s)
- Bo-Han Chen
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
| | - Chien-Chang Kao
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ting Xu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Ning Yang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tai-Lung Cha
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ta Tsai
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Liu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sheng-Tang Wu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - En Meng
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Wei Tsao
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Li Chen
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Guang-Huan Sun
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Dah-Shyong Yu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sun-Yran Chang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsin Yang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
57
|
Gorzo A, Galos D, Volovat SR, Lungulescu CV, Burz C, Sur D. Landscape of Immunotherapy Options for Colorectal Cancer: Current Knowledge and Future Perspectives beyond Immune Checkpoint Blockade. Life (Basel) 2022; 12:229. [PMID: 35207516 PMCID: PMC8878674 DOI: 10.3390/life12020229] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Colorectal cancer is the third most prevalent malignancy in Western countries and a major cause of death despite recent improvements in screening programs and early detection methods. In the last decade, a growing effort has been put into better understanding how the immune system interacts with cancer cells. Even if treatments with immune checkpoint inhibitors (anti-PD1, anti-PD-L1, anti-CTLA4) were proven effective for several cancer types, the benefit for colorectal cancer patients is still limited. However, a subset of patients with deficient mismatch repair (dMMR)/microsatellite-instability-high (MSI-H) metastatic colorectal cancer has been observed to have a prolonged benefit to immune checkpoint inhibitors. As a result, pembrolizumab and nivolumab +/- ipilimumab recently obtained the Food and Drug Administration approval. This review aims to highlight the body of knowledge on immunotherapy in the colorectal cancer setting, discussing the potential mechanisms of resistance and future strategies to extend its use.
Collapse
Affiliation(s)
- Alecsandra Gorzo
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania; (A.G.); (D.G.); (C.B.)
- Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400000 Cluj-Napoca, Romania
| | - Diana Galos
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania; (A.G.); (D.G.); (C.B.)
- Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400000 Cluj-Napoca, Romania
| | - Simona Ruxandra Volovat
- Department of Medical Oncology, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania;
| | | | - Claudia Burz
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania; (A.G.); (D.G.); (C.B.)
- Department of Allergology and Immunology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400000 Cluj-Napoca, Romania
| | - Daniel Sur
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania; (A.G.); (D.G.); (C.B.)
- Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400000 Cluj-Napoca, Romania
| |
Collapse
|
58
|
Jonasch E, Atkins MB, Chowdhury S, Mainwaring P. Combination of Anti-Angiogenics and Checkpoint Inhibitors for Renal Cell Carcinoma: Is the Whole Greater Than the Sum of Its Parts? Cancers (Basel) 2022; 14:cancers14030644. [PMID: 35158916 PMCID: PMC8833428 DOI: 10.3390/cancers14030644] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Checkpoint inhibitors and anti-angiogenic therapies are treatments that slow the progression of renal cell carcinoma, the most common type of kidney cancer. Checkpoint inhibitors and anti-angiogenic therapies work in different ways. Checkpoint inhibitors help to prevent tumors from hiding from the body’s immune system, while anti-angiogenic therapies slow the development of blood vessels that tumours need to help them to grow. Studies have shown that treatment with combination checkpoint inhibitor plus anti-angiogenic therapy can achieve better outcomes for patients with renal cell carcinoma than treatment with anti-angiogenic therapy alone. In this review, we consider how combination checkpoint inhibitor plus anti-angiogenic therapy works, and we review the current literature to identify evidence to inform clinicians as to the most effective way to use these different types of drugs, either one after the other, or together, for maximum patient benefit. Abstract Anti-angiogenic agents, such as vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitors and anti-VEGF antibodies, and immune checkpoint inhibitors (CPIs) are standard treatments for advanced renal cell carcinoma (aRCC). In the past, these agents were administered as sequential monotherapies. Recently, combinations of anti-angiogenic agents and CPIs have been approved for the treatment of aRCC, based on evidence that they provide superior efficacy when compared with sunitinib monotherapy. Here we explore the possible mechanisms of action of these combinations, including a review of relevant preclinical data and clinical evidence in patients with aRCC. We also ask whether the benefit is additive or synergistic, and, thus, whether concomitant administration is preferred over sequential monotherapy. Further research is needed to understand how combinations of anti-angiogenic agents with CPIs compare with CPI monotherapy or combination therapy (e.g., nivolumab and ipilimumab), and whether the long-term benefit observed in a subset of patients treated with CPI combinations will also be realised in patients treated with an anti-angiogenic therapy and a CPI. Additional research is also needed to establish whether other elements of the tumour microenvironment also need to be targeted to optimise treatment efficacy, and to identify biomarkers of response to inform personalised treatment using combination therapies.
Collapse
Affiliation(s)
- Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-792-2830
| | - Michael B. Atkins
- Department of Oncology, School of Medicine, Georgetown University, Washington, DC 20007, USA;
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| | - Simon Chowdhury
- Department of Medical Oncology, Guy’s and St Thomas’ Hospitals, London SE1 9RT, UK;
- Sarah Cannon Research Institute, London W1G 6AD, UK
| | - Paul Mainwaring
- Centre for Personalised Nanomedicine, The University of Queensland, Brisbane, QLD 4072, Australia;
| |
Collapse
|
59
|
Popovic M, Matovina-Brko G, Jovic M, Popovic LS. Immunotherapy: A new standard in the treatment of metastatic clear cell renal cell carcinoma. World J Clin Oncol 2022; 13:28-38. [PMID: 35116230 PMCID: PMC8790303 DOI: 10.5306/wjco.v13.i1.28] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 09/16/2021] [Accepted: 12/11/2021] [Indexed: 02/06/2023] Open
Abstract
Renal cell cancer (RCC) represents 2%-3% of all adulthood cancers and is the most common malignant neoplasm of the kidney (90%). In the mid-nineties of the last century, the standard of treatment for patients with metastatic RCC was cytokines. Sunititib and pazopanib were registered in 2007 and 2009, respectively, and have since been the standard first-line treatment for metastatic clear cell RCC (mccRCC). Renal cell cancer is a highly immunogenic tumor with tumor infiltrating cells, including CD8+ T lymphocytes, dendritic cells, natural killer cells (NK) and macrophages. This observation led to the design of new clinical trials in which patients were treated with immunotherapy. With the growing evidence that proangiogenic factors can have immunomodulatory effects on the host's immune system, the idea of combining angiogenic drugs with immunotherapy has emerged, and new clinical trials have been designed. In the last few years, several therapeutic options have been approved [immunotherapy and immunotherapy/tyrosine kinase inhibitors (TKI)] for the first-line treatment of mccRCC. Nivolumab/ipilimumab is approved for the treatment of patients with intermediate and poor prognoses. Several checkpoint inhibitors (pembrolizumab, nivolumab, avelumab) in combination with TKI (axitinib, lenvatinib, cabozantinib) are approved for the treatment of patients regardless of their International mRCC Database Consortium prognostic group and PD-L1 expression. There is no specific and ideal biomarker that could help in selecting the ideal patient for the appropriate first-line treatment.
Collapse
Affiliation(s)
- Maja Popovic
- Department of Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Sremska Kamenica 21204, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad 21000, Serbia
| | - Gorana Matovina-Brko
- Department of Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Sremska Kamenica 21204, Serbia
| | - Masa Jovic
- Department of Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Sremska Kamenica 21204, Serbia
| | - Lazar S Popovic
- Department of Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Sremska Kamenica 21204, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad 21000, Serbia
| |
Collapse
|
60
|
Maritaz C, Broutin S, Chaput N, Marabelle A, Paci A. Immune checkpoint-targeted antibodies: a room for dose and schedule optimization? J Hematol Oncol 2022; 15:6. [PMID: 35033167 PMCID: PMC8760805 DOI: 10.1186/s13045-021-01182-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-CTLA-4 and anti-PD-1/PD-L1 immune checkpoint inhibitors are therapeutic monoclonal antibodies that do not target cancer cells but are designed to reactivate or promote antitumor immunity. Dosing and scheduling of these biologics were established according to conventional drug development models, even though the determination of a maximum tolerated dose in the clinic could only be defined for anti-CTLA-4. Given the pharmacology of these monoclonal antibodies, their high interpatient pharmacokinetic variability, the actual clinical benefit as monotherapy that is observed only in a specific subset of patients, and the substantial cost of these treatments, a number of questions arise regarding the selected dose and the dosing interval. This review aims to outline the development of these immunotherapies and considers optimization options that could be used in clinical practice.
Collapse
Affiliation(s)
- Christophe Maritaz
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sophie Broutin
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Chaput
- Laboratory for Immunomonitoring in Oncology (LIO), Faculty of Pharmacy, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Unit (DITEP), LRTI U1015 INSERM, Gustave Roussy, Villejuif, France
| | - Angelo Paci
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France.
- Pharmacokinetic Unit, Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.
| |
Collapse
|
61
|
Vafaei S, Zekiy AO, Khanamir RA, Zaman BA, Ghayourvahdat A, Azimizonuzi H, Zamani M. Combination therapy with immune checkpoint inhibitors (ICIs); a new frontier. Cancer Cell Int 2022; 22:2. [PMID: 34980128 PMCID: PMC8725311 DOI: 10.1186/s12935-021-02407-8] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022] Open
Abstract
Recently, immune checkpoint inhibitors (ICIs) therapy has become a promising therapeutic strategy with encouraging therapeutic outcomes due to their durable anti-tumor effects. Though, tumor inherent or acquired resistance to ICIs accompanied with treatment-related toxicities hamper their clinical utility. Overall, about 60-70% of patients (e.g., melanoma and lung cancer) who received ICIs show no objective response to intervention. The resistance to ICIs mainly caused by alterations in the tumor microenvironment (TME), which in turn, supports angiogenesis and also blocks immune cell antitumor activities, facilitating tumor cells' evasion from host immunosurveillance. Thereby, it has been supposed and also validated that combination therapy with ICIs and other therapeutic means, ranging from chemoradiotherapy to targeted therapies as well as cancer vaccines, can capably compromise tumor resistance to immune checkpoint blocked therapy. Herein, we have focused on the therapeutic benefits of ICIs as a groundbreaking approach in the context of tumor immunotherapy and also deliver an overview concerning the therapeutic influences of the addition of ICIs to other modalities to circumvent tumor resistance to ICIs.
Collapse
Affiliation(s)
- Somayeh Vafaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Angelina O. Zekiy
- Department of Prosthetic Dentistry, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ramadhan Ado Khanamir
- Internal Medicine and Surgery Department, College of Veterinary Medicine, University of Duhok, Kurdistan Region, Iraq
| | - Burhan Abdullah Zaman
- Basic Sciences Department, College of Pharmacy, University of Duhok, Kurdistan Region, Iraq
| | | | | | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
62
|
Abdelrahim M, Abudayyeh A. Renal Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:389-397. [PMID: 34972976 DOI: 10.1007/978-3-030-79308-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
With the increasing use of immunotherapy, there has been an associated increased survival in many cancers but has also resulted in unregulated organ-specific toxicities. In this review, we will discuss the renal toxicities associated with a checkpoint inhibitor (CPI) from the typical acute tubulointerstitial nephritis to glomerulonephritis and their proposed mechanisms and treatments. We also discuss the use of CPI and reactivation of preexisting autoimmune disease with a focus on renal cell cancer in setting of chronic kidney disease (CKD). Transplant rejection in setting of CPI use has been further evaluated with single-center and multicenter retrospective studies, and available data will be presented in this chapter.
Collapse
Affiliation(s)
- Maen Abdelrahim
- Institute of Academic Medicine and Weill Cornell Medical College, Houston Methodist Cancer Center, Houston Methodist Cancer Center, Houston, TX, USA
| | - Ala Abudayyeh
- Division of Internal Medicine, Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
63
|
Targeting N6-methyladenosine RNA modification combined with immune checkpoint Inhibitors: A new approach for cancer therapy. Comput Struct Biotechnol J 2022; 20:5150-5161. [PMID: 36187919 PMCID: PMC9508382 DOI: 10.1016/j.csbj.2022.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022] Open
|
64
|
Kathuria-Prakash N, Drolen C, Hannigan CA, Drakaki A. Immunotherapy and Metastatic Renal Cell Carcinoma: A Review of New Treatment Approaches. Life (Basel) 2021; 12:24. [PMID: 35054417 PMCID: PMC8781308 DOI: 10.3390/life12010024] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Renal cell carcinomas (RCC) have been treated with immunotherapy for decades; the use of immune checkpoint inhibitors represents the most recent advance. In this review, we compare these new RCC immunotherapies, with a focus on achieving durable complete responses (CR). REVIEW Sorafenib and sunitinib were the first Food and Drug Administration (FDA)-approved targeted agents for RCC, with sunitinib eventually becoming the standard-of-care agent against which novel therapies are compared. In the last five years, many combination therapies based on the use of immune checkpoint inhibitors (ICIs) and receptor tyrosine kinase inhibitors (TKIs), including ipilimumab/nivolumab, nivolumab/cabozantinib, avelumab/axitinib, pembrolizumab/axitinib, and pembrolizumab/lenvatinib, have demonstrated superior overall survival (OS) and progression-free survival (PFS) compared to sunitinib. Ongoing clinical trials of hypoxia-induced factor-2 alpha (HIF-2a) inhibitors, chimeric antigen receptor T cell (CAR-T) therapy targeting CD70, and other new combination therapies have also shown promise and are currently under investigation. CONCLUSIONS Many new combination therapies are approved for RCC treatment, and CR rates suggest that, in the era of immunotherapy, it may be possible to achieve durable responses and survival benefit in patients with metastatic RCC.
Collapse
Affiliation(s)
- Nikhita Kathuria-Prakash
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (N.K.-P.); (C.D.); (C.A.H.)
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Claire Drolen
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (N.K.-P.); (C.D.); (C.A.H.)
| | - Christopher A. Hannigan
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (N.K.-P.); (C.D.); (C.A.H.)
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Alexandra Drakaki
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (N.K.-P.); (C.D.); (C.A.H.)
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
65
|
Navani V, Heng DYC. Treatment Selection in First-line Metastatic Renal Cell Carcinoma-The Contemporary Treatment Paradigm in the Age of Combination Therapy: A Review. JAMA Oncol 2021; 8:292-299. [PMID: 34792538 DOI: 10.1001/jamaoncol.2021.4337] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Importance The treatment landscape of metastatic renal cell carcinoma has evolved rapidly over the last decade. Recent combination approaches heralded by targeting immune checkpoints cytotoxic T-lymphocyte antigen 4 and programmed death-1 (PD-1) have been followed in consecutive years by protocols targeting vascular endothelial growth factor receptor, PD-1, and programmed death ligand-1. The differences in baseline patient characteristics, statistical plans, follow-up length, biomarker-derived approaches, and trial design make cross-trial comparisons difficult. Given the regulatory approval of a number of these regimens, the current available evidence is reviewed herein for combination first-line regimens with published randomized phase 3 trial data. Observations Combination approaches have transformed outcomes for patients. Durable disease control and prolonged overall survival have been achieved by both doublet immune checkpoint blockade and vascular endothelial growth factor receptor plus PD-1 blockade. Rationale for variations in trial outcome are offered, alongside approaches to navigating patient-empowered treatment selection, focusing on predictive tools, biomarkers, and the role of real-world data. Conclusions and Relevance Advances in the genomic, molecular, and immunologic understanding of metastatic clear cell renal cell carcinoma have lifted the survival curves for this disease markedly in recent years. Combination approaches will remain standard of care in the first-line setting. However, thoughtful study design is needed to accurately estimate outcomes and integrate novel approaches into the treatment armamentarium.
Collapse
Affiliation(s)
- Vishal Navani
- Medical Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Daniel Y C Heng
- Medical Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| |
Collapse
|
66
|
Liu Q, Yu Y, Lin J, Wang Y, Ai L, Li Q, Wu W, Jin H, Li S, Liu M, Yu S, Liu T. Treatment strategy for myocarditis in patients using immune checkpoint inhibitors or combined anti-vascular endothelial growth factor therapy by clinical severity. Eur J Cancer 2021; 157:10-20. [PMID: 34464781 DOI: 10.1016/j.ejca.2021.07.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/01/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Combination of immune checkpoint inhibitor (ICI) and anti-vascular endothelial growth factor (VEGF) therapy has increasingly become a promising strategy in various tumors. However, the combination might be associated with increased cardiotoxicities. Myocarditis is a potentially fatal complication in cancer patients treated with ICI. Currently, there are no clear guidelines for the management of this disease nor data characterizing the clinical course of myocarditis patients due to the combination of ICI and anti-VEGF therapy. PATIENTS AND METHODS This study included all patients consecutively admitted to Shanghai Zhongshan Hospital, Fudan University for the diagnosis of ICI-associated myocarditis according to Bonaca's criteria. The clinical presentation and outcome of myocarditis patients were collected receiving ICI and anti-VEGF combined therapy. RESULTS A total of 48 patients were included (23 received combined treatment of ICI and anti-VEGF while 25 received ICI only). No differences in baseline characteristics, clinical course, and outcomes were observed among patients receiving ICI with or without anti-VEGF treatment. The patients were subdivided into 3 groups including 8 fulminant cases, 25 clinically significant cases, and 15 subclinical cases based on clinical severity. The fulminant group was associated with a higher rate of cardiovascular deaths (CVDs) compared with clinically significant and subclinical groups (87.5% versus 4.0% versus 0.0%, p < 0.01). When stratified by the dose of corticosteroids used, cases with high-dose usage were more likely to have a CVD when compared to low dose or no use and intermediate dose groups (0.0% versus 4.0% versus 57.1%, p < 0.01). CONCLUSIONS No significant differences between myocarditis patients receiving ICI or combined anti-VEGF therapy in terms of clinical presentation and outcome were observed. Treatment strategy for myocarditis in patients using ICI or combined anti-VEGF therapy should be based on clinical severity. Specifically, immunosuppressive therapy besides high-dose corticosteroids is needed for fulminant cases.
Collapse
Affiliation(s)
- Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Jinyi Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Luoyan Ai
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Wei Wu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hang Jin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mengling Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Cancer Center, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
67
|
Mondlane ER, Abreu-Mendes P, Martins D, Cruz R, Mendes F. The role of immunotherapy in advanced renal cell carcinoma: Review. Int Braz J Urol 2021; 47:1228-1242. [PMID: 33650838 PMCID: PMC8486460 DOI: 10.1590/s1677-5538.ibju.2020.0681] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/05/2020] [Indexed: 01/05/2023] Open
Affiliation(s)
- Ercília Rita Mondlane
- ESTeSCPolitécnico de CoimbraCoimbraPortugalPolitécnico de Coimbra, ESTeSC, DFARM, ESTeSC, SM Bispo, Coimbra, Portugal.
| | - Pedro Abreu-Mendes
- Centro Hospital Universitário de São JoãoServiço de UrologiaPortoPortugalServiço de Urologia, Centro Hospital Universitário de São João, Porto, Portugal.
- Universidade do PortoFaculdade de MedicinaPortoPortugalFaculdade de Medicina Universidade do Porto, Porto, Portugal.
| | - Diana Martins
- ESTeSCPolitécnico de CoimbraCoimbraPortugalPolitécnico de Coimbra, ESTeSC, DCBL, SM Bispo, Coimbra, Portugal.
- Universidade de CoimbraInstituto de Investigação Clínica e Biomédica de Coimbra CoimbraPortugalUniversidade de Coimbra, Instituto de Investigação Clínica e Biomédica de Coimbra Coimbra, Portugal.
- Universidade de CoimbraCentro de Biomedicina e Biotecnologia Inovadoras (CIBB)CoimbraPortugalUniversidade de Coimbra, Centro de Biomedicina e Biotecnologia Inovadoras (CIBB), Coimbra, Portugal.
- Centro Académico Clínico de CoimbraCoimbraPortugalCentro Académico Clínico de Coimbra (CACC), Coimbra, Portugal.
- Universidade do PortoInstituto de Investigação e Inovação em SaúdePortoPortugalInstituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| | - Rui Cruz
- ESTeSCPolitécnico de CoimbraCoimbraPortugalPolitécnico de Coimbra, ESTeSC, DFARM, ESTeSC, SM Bispo, Coimbra, Portugal.
| | - Fernando Mendes
- ESTeSCPolitécnico de CoimbraCoimbraPortugalPolitécnico de Coimbra, ESTeSC, DCBL, SM Bispo, Coimbra, Portugal.
- Universidade de CoimbraInstituto de Investigação Clínica e Biomédica de Coimbra CoimbraPortugalUniversidade de Coimbra, Instituto de Investigação Clínica e Biomédica de Coimbra Coimbra, Portugal.
- Universidade de CoimbraCentro de Biomedicina e Biotecnologia Inovadoras (CIBB)CoimbraPortugalUniversidade de Coimbra, Centro de Biomedicina e Biotecnologia Inovadoras (CIBB), Coimbra, Portugal.
- Centro Académico Clínico de CoimbraCoimbraPortugalCentro Académico Clínico de Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
68
|
Tzeng A, Tzeng TH, Ornstein MC. Treatment-free survival after discontinuation of immune checkpoint inhibitors in metastatic renal cell carcinoma: a systematic review and meta-analysis. J Immunother Cancer 2021; 9:jitc-2021-003473. [PMID: 34599022 PMCID: PMC8488739 DOI: 10.1136/jitc-2021-003473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2021] [Indexed: 11/03/2022] Open
Abstract
While immune checkpoint inhibitors (ICI) can lead to sustained responses in metastatic renal cell carcinoma (mRCC), the optimal duration of therapy remains unknown. We aimed to examine treatment-free survival (TFS) in objective responders who discontinued ICI and to explore factors that may impact objective response rate (ORR) and TFS. MEDLINE/PubMed, Embase, and the Cochrane Library were searched for prospective studies reporting individual outcomes after ICI discontinuation in patients with mRCC. Pooled ORR and TFS were estimated using random-effects meta-analyses, and associations between ICI regimen type or treatment line and ORR or TFS were evaluated. Sixteen cohorts comprising 1833 patients treated with ICI were included. The pooled ORR was 43% (95% CI 33% to 53%), and significant differences in summary estimates existed among patients who received ICI monotherapy (22%, 95% CI 18% to 26%), ICI plus a vascular endothelial growth factor (VEGF) pathway inhibitor (57%, 95% CI 48% to 65%), and dual ICI (40%, 95% CI 36% to 44%). Of 572 responders who had available data, 327 stopped ICI, with 86 (26%) continuing to respond off-treatment. Pooled TFS rates at 6 and 12 months were 35% (95% CI 20% to 50%) and 20% (95% CI 8% to 35%), respectively, and were highest for responders treated with dual ICI and lowest for those treated with ICI plus a VEGF pathway inhibitor. Thus, a subset of patients with mRCC who are treated with ICI-based therapy can have durable TFS after therapy discontinuation. Prospective clinical trials and biomarkers are needed to identify patients who can discontinue ICI therapy without compromising clinical outcomes.
Collapse
Affiliation(s)
- Alice Tzeng
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tony H Tzeng
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Moshe C Ornstein
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland, Ohio, USA
| |
Collapse
|
69
|
Yuan M, Zang L, Xu A, Gong M, Liu Q, Huo B, Wang J, Fu H, Tse G, Roever L, Li G, Wang H, Liu T. Dynamic Changes of Serum Heart Type-Fatty Acid Binding Protein in Cancer Patients Treated With Immune Checkpoint Inhibitors. Front Pharmacol 2021; 12:748677. [PMID: 34658887 PMCID: PMC8517171 DOI: 10.3389/fphar.2021.748677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Objective: Immune checkpoint inhibitors (ICIs) are effective anti-cancer drugs that can improve survival in cancer patients, but their use may be associated with adverse cardiovascular side effects. Therefore, there is a clinical unmet need to identify non-invasive biomarker to detect subclinical cardiac toxicity after ICI treatment. The aim of this study is to examine the plasma levels of biomarkers in cancer survivors who were treated with ICIs. Patients and Methods: In a cohort of 19 cancer patients, biomarkers were evaluated at baseline, 1 month, 3 and 6 months after ICI administration. These biomarkers, hypothesized to be mechanistically relevant to cardiotoxicity, included cardiac troponin I (cTnI), high-sensitivity C-reactive protein (Hs-CRP), N-terminal pro-B-type natriuretic peptide (NT-pro BNP), CK (creatine kinase), CK-MB (creatine kinase-MB), Pentraxin-related protein 3 (PTX3), growth differentiation factor 15 (GDF-15), heart type-fatty acid binding protein (H-FABP) and galectin 3 (Gal-3). Results: H-FABP, but not other biomarkers, were increased at 3 months, which persisted at 6 months (529.28 ± 312.83 vs. 752.33 ± 283.65 vs. 808.00 ± 289.69 pg/ml, p = 0.031 and p = 0.013). Left ventricular ejection fraction (63.00 ± 4.15% vs. 63.74 ± 4.07%, p > 0.05) was not significantly reduced at this time point. Conclusions: H-FABP, but not other biomarkers, were increased in patients who were treated using ICIs. H-FABP might be a more sensitive biomarker to detect ICI-related subclinical myocardial damage than traditional cardiac biomarkers.
Collapse
Affiliation(s)
- Ming Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Zang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Aiqing Xu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Mengqi Gong
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qing Liu
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bin Huo
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
- Central Laboratory/Tianjin Research Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jinhuan Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huaying Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Leonardo Roever
- Department of Clinical Research, Federal University of Uberlândia, Uberlândia, Brazil
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haitao Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
- Central Laboratory/Tianjin Research Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
70
|
Eftekhar SP, Yazdanpanah N, Rezaei N. Immune checkpoint inhibitors and cardiotoxicity: possible mechanisms, manifestations, diagnosis and management. Expert Rev Anticancer Ther 2021; 21:1211-1228. [PMID: 34511008 DOI: 10.1080/14737140.2021.1979396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a new class of anticancer drugs that enhance the immune system function and activate T cells against cancerous cells. Although cardiac complications are not common, they could be accompanied with high morbidity and mortality. AREAS COVERED Regarding the importance of cardiac complications and their subsequent burden on individuals and the healthcare system, this review attempts to discuss the mechanism, diagnosis, and management of myocarditis, besides recapitulating the possible mechanism of other cardiac adverse events. Moreover, we briefly discuss the concurrent administration of other chemotherapeutic agents. EXPERT OPINION Due to insufficient knowledge concerning the physiopathology of immune-related adverse events (irAEs) and their potential further complications, cardiovascular complications in particular and in the context of this paper's focus, cooperation of oncologists, immunologists, and cardiologists is necessary for the management of patients. Experimental approaches such as using corticosteroids are becoming a part of guidelines for managing cardiac irAEs. However, a unique algorithm for diagnosis and management is necessary, especially in myocarditis cases. Furthermore, more studies are required to resolve current challenges, including prevention of myocarditis, concurrent administration of other chemotherapeutic agents, and re-introducing patients with ICIs.
Collapse
Affiliation(s)
- Seyed Parsa Eftekhar
- School of Medicine, Babol University of Medical Sciences, Babol, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Babol, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
71
|
Jin H, D'Urso V, Neuteboom B, McKenna SD, Schweickhardt R, Gross AW, Fomekong Nanfack Y, Paoletti A, Carter C, Toleikis L, Fluck M, Scheuenpflug J, Cai T. Avelumab internalization by human circulating immune cells is mediated by both Fc gamma receptor and PD-L1 binding. Oncoimmunology 2021; 10:1958590. [PMID: 34484871 PMCID: PMC8409756 DOI: 10.1080/2162402x.2021.1958590] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Avelumab is an IgG1 anti-programmed death ligand 1 (anti-PD-L1) monoclonal antibody that has been approved as a monotherapy for metastatic Merkel cell carcinoma and advanced urothelial carcinoma, and in combination with axitinib for advanced renal cell carcinoma. Avelumab is cleared faster and has a shorter half-life than other anti-PD-L1 antibodies, such as atezolizumab and durvalumab, but the mechanisms underlying these differences are unknown. IgG antibodies can be cleared through receptor-mediated endocytosis after binding of the antibody Fab region to target proteins, or via Fcγ receptor (FcγR)-mediated endocytosis. Unlike other approved anti-PD-L1 antibodies, avelumab has a native Fc region that retains FcγR binding capability. We hypothesized that the rapid clearance of avelumab might be due to the synergistic effect of both FcγR-mediated and PD-L1 target-mediated internalization. To investigate this, we performed in vitro and in vivo studies that compared engineered variants of avelumab and atezolizumab to determine mechanisms of cellular internalization. We found that both FcγR and PD-L1 binding contribute to avelumab internalization. While FcγR binding was the dominant mechanism of avelumab internalization in vitro, with CD64 acting as the most important FcγR, studies in mice and cynomolgus monkeys showed that both FcγR and PD-L1 contribute to avelumab elimination, with PD-L1 binding playing a greater role. These studies suggest that the rapid internalization of avelumab might be due to simultaneous binding of both PD-L1 and FcγR in trans. Our findings also provide a basis to alter the clearance and half-life of monoclonal antibodies in therapeutic development.
Collapse
Affiliation(s)
- Hulin Jin
- the healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Berend Neuteboom
- Istituto di Ricerche Biomediche "A. Marxer" RBM S.p.A. Colleretto Giacosa, Italy, an affiliate of Merck KGaA, Darmstadt, Germany
| | | | - Rene Schweickhardt
- Istituto di Ricerche Biomediche "A. Marxer" RBM S.p.A. Colleretto Giacosa, Italy, an affiliate of Merck KGaA, Darmstadt, Germany
| | | | | | - Andrea Paoletti
- Istituto di Ricerche Biomediche "A. Marxer" RBM S.p.A. Colleretto Giacosa, Italy, an affiliate of Merck KGaA, Darmstadt, Germany
| | | | - Lars Toleikis
- the healthcare business of Merck KGaA, Darmstadt, Germany
| | - Markus Fluck
- the healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Ti Cai
- EMD Serono, Billerica, MA, USA
| |
Collapse
|
72
|
Clearing up Clear Cell: Clarifying the Immuno-Oncology Treatment Landscape for Metastatic Clear Cell RCC. Cancers (Basel) 2021; 13:cancers13164140. [PMID: 34439293 PMCID: PMC8391664 DOI: 10.3390/cancers13164140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/28/2021] [Accepted: 08/13/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with advanced or malignant renal cell carcinoma at the time of diagnosis have historically had a poor prognosis. Immunonologic agents have significantly altered the therapeutic landscape and clinical outcomes of these patients. In this review, we highlight recent and upcoming clinical trials investigating the role of immunotherapies in clear cell RCC. In particular, we emphasize immunotherapy-based combinations, including immune checkpoint inhibitor (ICI) combinations, neoadjuvant, and adjuvant ICI, and ICI agents combined with anti-VEGF therapy.
Collapse
|
73
|
Conforti F, Marino M, Vitolo V, Spaggiari L, Mantegazza R, Zucali P, Ruffini E, di Tommaso L, Pelosi G, Barberis M, Petrini I, Palmieri G, Pasello G, Galli G, Berardi R, Garassino M, Filosso P, Alloisio M, Scorsetti M, Orecchia R, Pala L, Abatedaga L, Cinieri S, De Pas T. Clinical management of patients with thymic epithelial tumors: the recommendations endorsed by the Italian Association of Medical Oncology (AIOM). ESMO Open 2021; 6:100188. [PMID: 34116501 PMCID: PMC8193108 DOI: 10.1016/j.esmoop.2021.100188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/12/2021] [Accepted: 05/24/2021] [Indexed: 11/25/2022] Open
Abstract
The Italian Association of Medical Oncology recommendations on thymic epithelial tumors, which have been drawn up for the first time in 2020 through an evidence-based approach, report indications on all the main aspects of clinical management of this group of rare diseases, from diagnosis and staging, to new available systemic treatments, such as targeted therapies and immunotherapies. A summary of key recommendations is presented here and complete recommendations are reported as Supplementary Materials, available at https://doi.org/10.1016/j.esmoop.2021.100188.
Collapse
Affiliation(s)
- F Conforti
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, Istituto Europeo di Oncologia, IRCCS, Milan, Italy.
| | - M Marino
- Department of Pathological Anatomy, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - V Vitolo
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - L Spaggiari
- Department of Thoracic Surgery, IRCCS European Institute of Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - R Mantegazza
- Department of Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - P Zucali
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy
| | - E Ruffini
- Department of Thoracic Surgery, San Giovanni Battista Hospital, University of Torino, Torino, Italy
| | - L di Tommaso
- Department of Pathology, Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - G Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - M Barberis
- Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - I Petrini
- Department of Critical Area and Surgical, Medical and Molecular Pathology, University of Pisa, Pisa, Italy
| | - G Palmieri
- Scientific Direction, Department of Clinical Medicine and Surgery, Rare Tumors Reference Center, University Federico II, Naples, Italy
| | - G Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - G Galli
- Thoracic Oncology, Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - R Berardi
- Medical Oncology Unit, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I-GM Lancisi-G Salesi, Ancona, Italy
| | - M Garassino
- Thoracic Oncology, Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - P Filosso
- Thoracic Surgery Unit, University of Torino, Torino, Italy
| | - M Alloisio
- Department of Neurosurgery, Humanitas Clinical and Research Center-IRCSS, Rozzano, Italy
| | - M Scorsetti
- Department of Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center-IRCSS, Rozzano, Italy
| | - R Orecchia
- Scientific Direction, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - L Pala
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| | - L Abatedaga
- Associazione TU.TO.R. Tumori Toracici Rari, Milan, Italy
| | - S Cinieri
- Medical Oncology Unit, Antonio Perrino Hospital, Brindisi, Italy
| | - T De Pas
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| |
Collapse
|
74
|
Expression patterns and prognostic value of RUNX genes in kidney cancer. Sci Rep 2021; 11:14934. [PMID: 34294773 PMCID: PMC8298387 DOI: 10.1038/s41598-021-94294-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022] Open
Abstract
Kidney cancer is the third most common malignancy of the urinary system, of which, kidney renal clear cell carcinoma (KIRC) accounts for the vast majority. Runt-related transcription factors (RUNX) are involved in multiple cellular functions. However, the diverse expression patterns and prognostic values of RUNX genes in kidney cancer remained to be elucidated. In our study, we mined the DNA methylation, transcriptional and survival data of RUNX genes in patients with different kinds of kidney cancer through Oncomine, Gene Expression Profiling Interactive Analysis, UALCAN, Kaplan–Meier Plotter, cBioPortal and LinkedOmics. We found that RUNX1 and RUNX3 were upregulated in KIRC tissues compared with those in normal tissues. The survival analysis results indicated a high transcription level of RUNX1 was associated with poor overall survival (OS) in KIRC patients. Furthermore, KIRC tumor tissues had significantly lower levels of RUNX1 promoter methylation than that in paracancerous tissues, with decreased DNA methylation of RUNX1 notably associated with poor OS in KIRC. In conclusion, our results revealed that RUNX1 may be a potential therapeutic target for treating KIRC, and RUNX1 promoter methylation level shows promise as a novel diagnostic and prognostic biomarker, which laid a foundation for further study.
Collapse
|
75
|
Wang S, Chen S, Ying Y, Ma X, Shen H, Li J, Wang X, Lin Y, Liu B, Zheng X, Xie L. Comprehensive Analysis of Ferroptosis Regulators With Regard to PD-L1 and Immune Infiltration in Clear Cell Renal Cell Carcinoma. Front Cell Dev Biol 2021; 9:676142. [PMID: 34291048 PMCID: PMC8287329 DOI: 10.3389/fcell.2021.676142] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/03/2021] [Indexed: 01/05/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the tumor types with sensitivity to ferroptosis, and immunotherapy has emerged as a standard pillar for metastatic ccRCC treatment, while it remains largely obscure whether ferroptosis influences the tumor immune microenvironment in ccRCC. Based on available data in The Cancer Genome Atlas, divergent expression profiles of ferroptosis regulators were noted in ccRCC and normal tissues, and we also found that the ferroptosis regulators correlated with the PD-L1 expression. Two independent subtypes were determined by consensus clustering analysis according to the expression level of ferroptosis regulators in ccRCC. Cluster 1 showed lower histological tumor stage and grade, more favorable prognosis, and higher PD-L1 expression compared to cluster 2. CIBERSORT analysis revealed that cluster 2 harbored higher infiltrated levels of CD8+ T cell, Tregs, and T follicular helper cell, while cluster 1 more correlated with the monocyte, M1 macrophage, and M2 macrophage. Gene set enrichment analysis indicated that the ERBB signaling and JAK_STAT signaling pathways were significantly enriched in cluster 1. We subsequently identified CARS as the potentially key immune infiltration-related ferroptosis regulator, whose high expression showed dismal prognosis and was positively correlated with PD-L1 expression in ccRCC. We also verified the upregulation of CARS in ccRCC tissues and cell lines via qRT-PCR method. Additionally, a pan-cancer analysis demonstrated that CARS closely related to the expression of immune checkpoint-related genes (especially PD-L1) and an unfavorable prognosis in diverse cancer types. In summary, our study suggested the crucial role of ferroptosis in immune infiltration of ccRCC, and CARS is a potentially novel prognostic biomarker and potential target for cancer immunotherapy.
Collapse
Affiliation(s)
- Song Wang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiming Chen
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yufan Ying
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xueyou Ma
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haixiang Shen
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiangfeng Li
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Wang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiwei Lin
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ben Liu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiangyi Zheng
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liping Xie
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
76
|
Wang S, Ma X, Ying Y, Sun J, Yang Z, Li J, Jin K, Wang X, Xie B, Zheng X, Liu B, Xie L. Upregulation of ARNTL2 is associated with poor survival and immune infiltration in clear cell renal cell carcinoma. Cancer Cell Int 2021; 21:341. [PMID: 34217271 PMCID: PMC8255002 DOI: 10.1186/s12935-021-02046-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Background Aryl hydrocarbon receptor nuclear translocator like 2 (ARNTL2) is a member of the PAS superfamily. Previous studies explored the carcinogenic roles of transcription factor ARNTL2 in human malignancies. However, its roles in ccRCC have not been elucidated. This study sought to explore the roles of ARNTL2 in ccRCC and determine its correlations with tumor immunity. Methods The expression of ARNTL2 was analyzed using the GEO, TCGA and GTEx database, and verified in ccRCC tissue samples and cell lines by qRT-PCR and western blot analysis. Kaplan–Meier survival curve analysis, Cox regression analysis (including univariate and multivariate analysis) was utilized to evaluate the prognostic values of ARNTL2. Potential biological mechanisms of ARNTL2 were explored using GSEA method. Colony formation and wound healing assays were conducted to explore the oncogenic role of ARNTL2 in ccRCC. ssGSEA and xCell algorithm were used to explore the correlation between ARNTL2 expression and tumor immune microenvironment (TIME). Results ARNTL2 was significantly upregulated in ccRCC tissues and cell lines compared to normal kidney tissues and cell line. Enhanced expression of ARNTL2 was strongly linked to advanced clinical stage and unfavorable overall survival in ccRCC. ARNTL2 was determined as an independent prognostic marker through cox regression analysis. A prognostic nomogram was constructed to predict 1-, 3- and 5-year overall survival of ccRCC patients by integrating ARNTL2 expression with other clinicopathologic variables. GSEA analysis showed that focal adhesion, T cell receptor, cell cycle, and JAK-STAT signaling pathway were significantly enriched in high ARNTL2 samples. Silencing of ARNTL2 suppressed the colony formation ability and wound healing efficacy of ccRCC cell lines. xCell analysis showed that high expression level of ARNTL2 exhibited an immune infiltration status similar to CD8 + inflamed ccRCC subtype, which was characterized by high infiltration level of CD8 + T cell and high expression level of the immune escape biomarkers such as PD-L1, PD-L2, PD1 and CTLA4. Conclusion ARNTL2 is an independent adverse predictor of ccRCC patient survival. High expression level of ARNTL2 is associated with immune infiltration, and may be a novel therapeutic target in ccRCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02046-z.
Collapse
Affiliation(s)
- Song Wang
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Xueyou Ma
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Yufan Ying
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jiazhu Sun
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Zitong Yang
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jiangfeng Li
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Ke Jin
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Xiao Wang
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Bo Xie
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Xiangyi Zheng
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Ben Liu
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Liping Xie
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
77
|
Tan AC, Bagley SJ, Wen PY, Lim M, Platten M, Colman H, Ashley DM, Wick W, Chang SM, Galanis E, Mansouri A, Khagi S, Mehta MP, Heimberger AB, Puduvalli VK, Reardon DA, Sahebjam S, Simes J, Antonia SJ, Berry D, Khasraw M. Systematic review of combinations of targeted or immunotherapy in advanced solid tumors. J Immunother Cancer 2021; 9:jitc-2021-002459. [PMID: 34215688 PMCID: PMC8256733 DOI: 10.1136/jitc-2021-002459] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2021] [Indexed: 01/02/2023] Open
Abstract
With rapid advances in our understanding of cancer, there is an expanding number of potential novel combination therapies, including novel-novel combinations. Identifying which combinations are appropriate and in which subpopulations are among the most difficult questions in medical research. We conducted a Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)-guided systematic review of trials of novel-novel combination therapies involving immunotherapies or molecular targeted therapies in advanced solid tumors. A MEDLINE search was conducted using a modified Cochrane Highly Sensitive Search Strategy for published clinical trials between July 1, 2017, and June 30, 2020, in the top-ranked medical and oncology journals. Trials were evaluated according to a criterion adapted from previously published Food and Drug Administration guidance and other key considerations in designing trials of combinations. This included the presence of a strong biological rationale, the use of a new established or emerging predictive biomarker prospectively incorporated into the clinical trial design, appropriate comparator arms of monotherapy or supportive external data sources and a primary endpoint demonstrating a clinically meaningful benefit. Of 32 identified trials, there were 11 (34%) trials of the novel-novel combination of anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) therapy, and 10 (31%) trials of anti-PD-1/PD-L1 and anti-vascular endothelial growth factor (VEGF) combination therapy. 20 (62.5%) trials were phase II trials, while 12 (37.5%) were phase III trials. Most (72%) trials lacked significant preclinical evidence supporting the development of the combination in the given indication. A majority of trials (69%) were conducted in biomarker unselected populations or used pre-existing biomarkers within the given indication for patient selection. Most studies (66%) were considered to have appropriate comparator arms or had supportive external data sources such as prior studies of monotherapy. All studies were evaluated as selecting a clinically meaningful primary endpoint. In conclusion, designing trials to evaluate novel-novel combination therapies presents numerous challenges to demonstrate efficacy in a comprehensive manner. A greater understanding of biological rationale for combinations and incorporating predictive biomarkers may improve effective evaluation of combination therapies. Innovative statistical methods and increasing use of external data to support combination approaches are potential strategies that may improve the efficiency of trial design. Designing trials to evaluate novel-novel combination therapies presents numerous challenges to demonstrate efficacy in a comprehensive manner. A greater understanding of biological rationale for combinations and incorporating predictive biomarkers may improve effective evaluation of combination therapies. Innovative statistical methods and increasing use of external data to support combination approaches are potential strategies that may improve the efficiency of trial design.
Collapse
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,Duke-NUS Medical School, National University of Singapore, Singapore
| | - Stephen J Bagley
- Abramson Cancer Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Stanford, California, USA
| | - Michael Platten
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Centre, Heidelberg, Germany
| | - Howard Colman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - David M Ashley
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Wolfgang Wick
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Evanthia Galanis
- Division of Medical Oncology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Simon Khagi
- Division of Medical Oncology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida, USA
| | - Amy B Heimberger
- Department of Neurosurgery, Northwestern University, Chicago, Illinois, USA
| | - Vinay K Puduvalli
- Department of Neurooncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Solmaz Sahebjam
- Department of Neuro-oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Scott J Antonia
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Don Berry
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mustafa Khasraw
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
78
|
Kudo M, Motomura K, Wada Y, Inaba Y, Sakamoto Y, Kurosaki M, Umeyama Y, Kamei Y, Yoshimitsu J, Fujii Y, Aizawa M, Robbins PB, Furuse J. Avelumab in Combination with Axitinib as First-Line Treatment in Patients with Advanced Hepatocellular Carcinoma: Results from the Phase 1b VEGF Liver 100 Trial. Liver Cancer 2021; 10:249-259. [PMID: 34239811 PMCID: PMC8237783 DOI: 10.1159/000514420] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/13/2021] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Combining an immune checkpoint inhibitor with a targeted antiangiogenic agent may leverage complementary mechanisms of action for the treatment of advanced/metastatic hepatocellular carcinoma (aHCC). Avelumab is a human anti-PD-L1 IgG1 antibody with clinical activity in various tumor types; axitinib is a selective tyrosine kinase inhibitor of vascular endothelial growth factor receptors 1, 2, and 3. We report the final analysis from VEGF Liver 100 (NCT03289533), a phase 1b study evaluating safety and efficacy of avelumab plus axitinib in treatment-naive patients with aHCC. METHODS Eligible patients had confirmed aHCC, no prior systemic therapy, ≥1 measurable lesion, Eastern Cooperative Oncology Group performance status ≤1, and Child-Pugh class A disease. Patients received avelumab 10 mg/kg intravenously every 2 weeks plus axitinib 5 mg orally twice daily until progression, unacceptable toxicity, or withdrawal. Endpoints included safety and investigator-assessed objective response per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 and modified RECIST (mRECIST) for HCC. RESULTS Twenty-two Japanese patients were enrolled and treated with avelumab plus axitinib. The minimum follow-up was 18 months as of October 25, 2019 (data cutoff). Grade 3 treatment-related adverse events (TRAEs) occurred in 16 patients (72.7%); the most common (≥3 patients) were hypertension (n = 11 [50.0%]), palmar-plantar erythrodysesthesia syndrome (n = 5 [22.7%]), and decreased appetite (n = 3 [13.6%]). No grade 4 TRAEs or treatment-related deaths occurred. Ten patients (45.5%) had an immune-related AE (irAE) of any grade; 3 patients (13.6%) had an infusion-related reaction (IRR) of any grade, and no grade ≥3 irAE and IRR were observed. The objective response rate was 13.6% (95% CI: 2.9-34.9%) per RECIST 1.1 and 31.8% (95% CI: 13.9-54.9%) per mRECIST for HCC. CONCLUSION Treatment with avelumab plus axitinib was associated with a manageable toxicity profile and showed antitumor activity in patients with aHCC.
Collapse
Affiliation(s)
- Masatoshi Kudo
- Faculty of Medicine, Kindai University, Osaka, Japan,*Masatoshi Kudo,
| | | | | | | | | | | | | | | | | | | | | | | | - Junji Furuse
- Faculty of Medicine, Kyorin University, Tokyo, Japan
| |
Collapse
|
79
|
Individualizing renal cell carcinoma treatment through biomarkers discovery in the era of immune checkpoint inhibitors: where do we stand? Curr Opin Urol 2021; 31:236-241. [PMID: 33742982 DOI: 10.1097/mou.0000000000000870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The treatment landscape of metastatic renal cell carcinoma has greatly evolved over the past fifteen years, leading to a significant improvement in the outcome of our patients. However, there is still an urgent need for predictive biomarkers that could guide our treatment selection, especially in the present era of immune-based treatments. RECENT FINDINGS A number of putative biomarkers of immunotherapy activity have been proposed over the past few years, including PD-L1 immunohistochemical expression, tumor mutational burden, neoantigens load, insertions and deletions, complex gene signatures, as well as lymphocytic subpopulations (either circulating or tumor-infiltrating). However, despite preliminary intriguing findings, no biomarker for immune checkpoint activity has emerged so far, that could be used in everyday clinical practice, mainly due to preliminary, or frankly, conflicting results. SUMMARY The quest for an 'ideal' biomarker, which should be characterized by adequate specificity, sensibility, predictive (and not just prognostic) value, robustness, reproducibility, ease of evaluation and low cost, is still ongoing.
Collapse
|
80
|
Jacob JB, Jacob MK, Parajuli P. Review of immune checkpoint inhibitors in immuno-oncology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 91:111-139. [PMID: 34099106 DOI: 10.1016/bs.apha.2021.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tumor cells predominantly express self-antigens and overcoming self-tolerance is the primary challenge to effective immunotherapy. Tumors also express ligands for co-inhibitory molecules on immune cells, in order to suppress anti-tumor immunity. Over a decade ago, the first antibodies generated to block the co-inhibitory molecule CTLA-4 was tested in patients with metastatic melanoma. Results from this landmark trial have informed not only the current landscape of checkpoint blockade but also the way in which immunotherapy trial outcomes are determined. Antibodies targeting PD-1 and its ligand, PD-L1, soon followed and use of these checkpoint inhibitors (ICIs) have expanded exponentially. ICI treatment has shown long-lasting clinical benefit in several tumor types and patients refractory to other treatments can often respond to ICI therapy. On the other hand, in some tumor types, the response to ICI is short-lived and tumors eventually recur. Current clinical trials are focused on enhancing anti-tumor effects through combinations of multiple ICIs with agents which cause tumor death, particularly in solid tumors, in order to enhance antigen presentation. It is also important to define which patients will respond to therapy with ICIs as over half of all patients suffer from immune-related adverse events (irAE), some of which are severe and long-lasting.
Collapse
Affiliation(s)
- Jennifer B Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States.
| | - Mark K Jacob
- Michigan State University School of Human Medicine, East Lansing, MI, United States
| | - Prahlad Parajuli
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
81
|
Zhang Z, Xiong L, Wu Z, Liu H, Ning K, Peng Y, Yu C, Ding Y, Weng D, Xia J, Jiang L, Guo S, Han H, Zhou F, Dong P. Neoadjuvant combination of pazopanib or axitinib and programmed cell death protein-1-activated dendritic cell-cytokine-induced killer cells immunotherapy may facilitate surgery in patients with renal cell carcinoma. Transl Androl Urol 2021; 10:2091-2102. [PMID: 34159090 PMCID: PMC8185689 DOI: 10.21037/tau-21-406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background Radical/cytoreductive nephrectomy or nephron-sparing surgery may be thought to be not safe or unfeasible in some renal cell carcinoma (RCC) patients in which tumor is locally advanced or highly complicated. Neoadjuvant therapy may reduce the volume of the tumor, thus facilitates surgery. The aim the study is to evaluate the efficacy and safety of neoadjuvant combination of pazopanib or axitinib and PD-1-activated dendritic cell-cytokine-induced killer (PD-1/DC-CIK) cell immunotherapy in those patients. Methods Data from 16 RCC patients who received neoadjuvant pazopanib (Group P, n=9) or axitinib (Group A, n=7) plus PD-1/DC-CIK cells immunotherapy were reviewed retrospectively. A total of 9 participants that were potential candidates for radical/cytoreductive nephrectomy (RN/CN) had locally advanced tumor and 5 participants with partial nephrectomy (PN) absolute indications had highly complicated tumors. The efficacy outcomes were based on volume changes of the primary tumor, lymph nodes, and tumor thrombus in 13 participants with complete computed tomography (CT) imaging. The treatment-related toxicities and surgical complications were also reported. Results With a median of 2.1 months treatment, the overall volume of the tumors decreased by a median of 42.30% [interquartile range (IQR): 19.37–66.78%]. Specifically, the median reduction of tumor volume was 88.77 and 15.50 cm3 in group P and group A, respectively (P=0.014). However, participants in Group P were more likely to experience grade 3 or 4 treatment-related adverse events (AEs) than those in Group A (44.4% vs. 0). Finally, all participants were candidates for appropriate surgery after neoadjuvant therapy (as assessed by the surgeon), and 10 participants accepted surgery, including 5 PN, 4 RN/CN, and 1 lymph node dissection. A solitary participant had Clavien grade IV acute renal failure required dialysis and another had grade II lymphatic leakage. Conclusions Neoadjuvant combination of pazopanib or axitinib and PD-1/DC-CIK cells immunotherapy was well-tolerated and could effectively reduce the volume of tumors in locally advanced or highly complicated RCC patients.
Collapse
Affiliation(s)
- Zhiling Zhang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Longbin Xiong
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zeshen Wu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huiming Liu
- State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kang Ning
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yulu Peng
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chunping Yu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ya Ding
- State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Desheng Weng
- State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianchuan Xia
- State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lijuan Jiang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shengjie Guo
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hui Han
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Pei Dong
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
82
|
A Phase I/II Study to Assess the Safety and Efficacy of Pazopanib and Pembrolizumab Combination Therapy in Patients with Advanced Renal Cell Carcinoma. Clin Genitourin Cancer 2021; 19:434-446. [PMID: 34006498 DOI: 10.1016/j.clgc.2021.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND This study assessed whether antiangiogenic treatment may potentiate immune checkpoint blockade in patients with advanced renal cell carcinoma. PATIENTS AND METHODS This was an open-label, two-part, multicenter study involving treatment-naïve patients with advanced renal cell carcinoma. Part 1 consisted of a phase I dose escalation and expansion of pazopanib plus pembrolizumab (combination therapy). Cohorts A and B received pazopanib in combination with pembrolizumab, whereas Cohort C received pazopanib monotherapy for 9 weeks before receiving the combination therapy. Part 2 was planned as a randomized three-arm study but was not conducted. RESULTS Overall, 42 patients were enrolled (10 each in Cohorts A and B, 22 in Cohort C). The maximum tolerated dose was not reached and the recommended phase II dose was not declared, as Cohort C was closed early because of safety concerns. The overall response rates were 60% and 20% in Cohorts A and B, respectively. In Cohort C, the overall response rates were 33%, 25%, and 0% in the combination therapy, pembrolizumab monotherapy, and pazopanib monotherapy groups, respectively. The median progression-free survival rates were 21.95 months and 41.40 months in Cohorts A and B, respectively. Grade 3 or 4 adverse events (AEs) were observed in 90% of patients in Cohorts A and B. In Cohort C, the frequencies of grade 3 or 4 AEs, serious adverse events, and AEs leading to dose reduction were typically high in the combination therapy group. CONCLUSIONS Despite preliminary signs of efficacy, significant hepatotoxicity was observed in Cohorts A and B. The sequential schedule of pazopanib followed by pazopanib plus pembrolizumab showed reduced hepatotoxicity; however, other safety issues emerged with this approach.
Collapse
|
83
|
Rini BI, Atkins MB, Choueiri TK, Thomaidou D, Rosbrook B, Thakur M, Hutson TE. Time to Resolution of Axitinib-Related Adverse Events After Treatment Interruption in Patients With Advanced Renal Cell Carcinoma. Clin Genitourin Cancer 2021; 19:e306-e312. [PMID: 33947608 DOI: 10.1016/j.clgc.2021.03.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/19/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Combined axitinib and immuno-oncology (IO) therapy is approved for first-line advanced renal cell carcinoma. Overlapping toxicities represent a clinical challenge. Calculating the time to resolution (TTR) of common axitinib-related adverse events (AEs) after treatment interruption may help to identify AE etiology and determine appropriate management strategies. MATERIALS AND METHODS Data from 5 randomized or single-arm axitinib monotherapy or combination studies were analyzed. Patients with histologically confirmed clear cell advanced renal cell carcinoma were pooled into 3 cohorts based on treatment received: axitinib monotherapy, axitinib + IO, and other tyrosine kinase inhibitor (TKI). Any grade and grade ≥3 treatment-emergent diarrhea, fatigue, hypertension, nausea, and palmar-plantar erythrodysesthesia syndrome were assessed. TTR was defined as the time from treatment interruption/discontinuation to resolution. RESULTS The axitinib monotherapy cohort comprised 532 patients, the axitinib + IO cohort 541 patients, and the other TKI cohort 882 patients. Median TTR for all AEs (any grade) in the axitinib monotherapy cohort ranged from 1 to 3 days, except for fatigue (8 days). For diarrhea, hypertension, nausea, and palmar-plantar erythrodysesthesia syndrome, median TTRs were longer in the axitinib + IO (4-11 days) and other TKI (7-8 days) cohorts versus the monotherapy cohort. Results were similar when only AEs of grade ≥3 were considered. CONCLUSIONS The TTR of monotherapeutic axitinib-related AEs is ≤3 days, except for fatigue, and generally shorter than for other single-agent TKIs and axitinib + IO. This has important implications for identifying AE etiology with combined axitinib-IO therapy and implementation of appropriate management strategies. ClinicalTrials.org identifiers: NCT00678392, NCT00920816, NCT02493751, NCT02684006, NCT02853331.
Collapse
Affiliation(s)
- Brian I Rini
- Division of Hematology/Oncology, Vanderbilt University Medical Center, and Vanderbilt-Ingram Cancer Center, Nashville, TN.
| | | | | | | | - Brad Rosbrook
- Pfizer Global Product Development-Oncology, San Diego, CA 92121
| | | | | |
Collapse
|
84
|
Jang A, Sweeney PL, Barata PC, Koshkin VS. PD-L1 Expression and Treatment Implications in Metastatic Clear Cell Renal Cell Carcinoma: A Systematic Review. KIDNEY CANCER 2021. [DOI: 10.3233/kca-200108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Over the past decade, immune checkpoint inhibitors (ICIs) have increasingly become the standard of care for various advanced malignancies, including metastatic clear cell renal cell carcinoma (mccRCC). Most ICIs currently used in clinical practice inhibit the interaction between the programmed cell death protein-1 (PD-1) and programmed death ligand-1 (PD-L1) complex. A deeper understanding of this interaction and PD-L1 expression in tumors has led to more effective therapies in the treatment of advanced cancers, but the debate regarding the utility of PD-L1 as a biomarker continues. OBJECTIVE: We aimed to systematically evaluate the role of PD-L1 in mccRCC in terms of expression and treatment implications. METHODS: Following PRISMA guidelines, we performed a systematic literature search using PubMed and Embase through August 31, 2020. Titles and abstracts were screened to identify articles for full-text review. A hand search was also performed using Google Scholar and the bibliography to relevant studies. RESULTS: A total of 26 articles were identified, and relevant data were extracted and organized. The available information regarding PD-L1 expression in mccRCC from both prospective clinical trials and retrospective studies were summarized. We discussed the utility of PD-L1 as a predictive and prognostic biomarker in mccRCC, its association with other potential biomarkers, and the pattern and level of expression of PD-L1 in primary versus metastatic tumors. CONCLUSIONS: Although significant progress has been made, much more remains to be learned regarding the differences between PD-L1+ and PD-L1- ccRCC tumors, in terms of both the underlying biology and clinical responses to immunotherapy and other agents.
Collapse
Affiliation(s)
- Albert Jang
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Patrick L. Sweeney
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Pedro C. Barata
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center, New Orleans, LA, USA
| | - Vadim S. Koshkin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
85
|
Jian Y, Yang K, Sun X, Zhao J, Huang K, Aldanakh A, Xu Z, Wu H, Xu Q, Zhang L, Xu C, Yang D, Wang S. Current Advance of Immune Evasion Mechanisms and Emerging Immunotherapies in Renal Cell Carcinoma. Front Immunol 2021; 12:639636. [PMID: 33767709 PMCID: PMC7985340 DOI: 10.3389/fimmu.2021.639636] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
Renal cell carcinoma is a highly heterogeneous cancer group, and the complex microenvironment of the tumor provides appropriate immune evasion opportunities. The molecular mechanism of immune escape in renal cell carcinoma is currently a hot issue, focusing primarily on the major complex of histocompatibility, immunosuppressive cells, their secreted immunosuppressive cytokines, and apoptosis molecule signal transduction. Immunotherapy is the best treatment option for patients with metastatic or advanced renal cell carcinoma and combination immunotherapy based on a variety of principles has shown promising prospects. Comprehensive and in-depth knowledge of the molecular mechanism of immune escape in renal cell carcinoma is of vital importance for the clinical implementation of effective therapies. The goal of this review is to address research into the mechanisms of immune escape in renal cell carcinoma and the use of the latest immunotherapy. In addition, we are all looking forward to the latest frontiers of experimental combination immunotherapy.
Collapse
Affiliation(s)
- Yuli Jian
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Kangkang Yang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Xiaoxin Sun
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Jun Zhao
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kai Huang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Abdullah Aldanakh
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhongyang Xu
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Haotian Wu
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiwei Xu
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Chunyan Xu
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Deyong Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| |
Collapse
|
86
|
Sivaccumar J, Sandomenico A, Vitagliano L, Ruvo M. Monoclonal Antibodies: A Prospective and Retrospective View. Curr Med Chem 2021; 28:435-471. [PMID: 32072887 DOI: 10.2174/0929867327666200219142231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Monoclonal Antibodies (mAbs) represent one of the most important classes of biotherapeutic agents. They are used to cure many diseases, including cancer, autoimmune diseases, cardiovascular diseases, angiogenesis-related diseases and, more recently also haemophilia. They can be highly varied in terms of format, source, and specificity to improve efficacy and to obtain more targeted applications. This can be achieved by leaving substantially unchanged the basic structural components for paratope clustering. OBJECTIVES The objective was to trace the most relevant findings that have deserved prestigious awards over the years, to report the most important clinical applications and to emphasize their latest emerging therapeutic trends. RESULTS We report the most relevant milestones and new technologies adopted for antibody development. Recent efforts in generating new engineered antibody-based formats are briefly reviewed. The most important antibody-based molecules that are (or are going to be) used for pharmacological practice have been collected in useful tables. CONCLUSION The topics here discussed prove the undisputed role of mAbs as innovative biopharmaceuticals molecules and as vital components of targeted pharmacological therapies.
Collapse
Affiliation(s)
- Jwala Sivaccumar
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Luigi Vitagliano
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| |
Collapse
|
87
|
Li F, Jin Y, Pei X, Guo P, Dong K, Wang H, Chen Y, Guo P, Meng LB, Wang Z. Bioinformatics analysis and verification of gene targets for renal clear cell carcinoma. Comput Biol Chem 2021; 92:107453. [PMID: 33636636 DOI: 10.1016/j.compbiolchem.2021.107453] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/09/2020] [Accepted: 02/05/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND It is estimated that there are 338,000 new renal-cell carcinoma releases every year in the world. Renal cell carcinoma (RCC) is a heterogeneous tumor, of which more than 70% is clear cell renal cell carcinoma (ccRCC). It is estimated that about 30% of new renal-cell carcinoma patients have metastases at the time of diagnosis. However, the pathogenesis of renal clear cell carcinoma has not been elucidated. Therefore, it is necessary to further study the pathogenesis of ccRCC. METHODS Two expression profiling datasets (GSE68417, GSE71963) were downloaded from the GEO database. Differentially expressed genes (DEGs) between ccRCC and normal tissue samples were identified by GEO2R. Functional enrichment analysis was made by the DAVID tool. Protein-protein interaction (PPI) network was constructed. The hub genes were excavated. The clustering analysis of expression level of hub genes was performed by UCSC (University of California Santa Cruz) Xena database. The hub gene on overall survival rate (OS) in patients with ccRCC was performed by Kaplan-Meier Plotter. Finally, we used the ccRCC renal tissue samples to verify the hub genes. RESULTS 1182 common DEGs between the two datasets were identified. The results of GO and KEGG analysis revealed that variations in were predominantly enriched in intracellular signaling cascade, oxidation reduction, intrinsic to membrane, integral to membrane, nucleoside binding, purine nucleoside binding, pathways in cancer, focal adhesion, cell adhesion molecules. 10 hub genes ITGAX, CD86, LY86, TLR2, TYROBP, FCGR2A, FCGR2B, PTPRC, ITGB2, ITGAM were identified. FCGR2B and TYROBP were negatively correlated with the overall survival rate in patients with ccRCC (P < 0.05). RT-qPCR analysis showed that the relative expression levels of CD86, FCGR2A, FCGR2B, TYROBP, LY86, and TLR2 were significantly higher in ccRCC samples, compared with the adjacent renal tissue groups. CONCLUSIONS In summary, bioinformatics technology could be a useful tool to predict the progression of ccRCC. In addition, there are DEGs between ccRCC tumor tissue and normal renal tissue, and these DEGs might be considered as biomarkers for ccRCC.
Collapse
Affiliation(s)
- Feng Li
- Department of Urology, The Fourth Hospital of Hebei Medical University, No.12 Jiankang Road Shijiazhuang, 050011, Hebei Province, China.
| | - Yi Jin
- Department of Oncology, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, 054001, Hebei Province, China.
| | - Xiaolu Pei
- Department of Oncology, The Heibei General Hospital, No.348 Heping Road Shijiazhuang, 050051, Hebei Province, China.
| | - Peiyuan Guo
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, China.
| | - Keqin Dong
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, China.
| | - Haoyuan Wang
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, China.
| | - Yujia Chen
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, China.
| | - Peng Guo
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, No.12 Jiankang Road Shijiazhuang, 050011, Hebei Province, China.
| | - Ling-Bing Meng
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, China.
| | - Zhiyu Wang
- Department of Immuno-oncology, The Fourth Hospital of Hebei Medical University, No.12 Jiankang Road Shijiazhuang, 050011, Hebei Province, China.
| |
Collapse
|
88
|
Borcherding N, Vishwakarma A, Voigt AP, Bellizzi A, Kaplan J, Nepple K, Salem AK, Jenkins RW, Zakharia Y, Zhang W. Mapping the immune environment in clear cell renal carcinoma by single-cell genomics. Commun Biol 2021; 4:122. [PMID: 33504936 PMCID: PMC7840906 DOI: 10.1038/s42003-020-01625-6] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/17/2020] [Indexed: 01/30/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most immunologically distinct tumor types due to high response rate to immunotherapies, despite low tumor mutational burden. To characterize the tumor immune microenvironment of ccRCC, we applied single-cell-RNA sequencing (SCRS) along with T-cell-receptor (TCR) sequencing to map the transcriptomic heterogeneity of 25,688 individual CD45+ lymphoid and myeloid cells in matched tumor and blood from three patients with ccRCC. We also included 11,367 immune cells from four other individuals derived from the kidney and peripheral blood to facilitate the identification and assessment of ccRCC-specific differences. There is an overall increase in CD8+ T-cell and macrophage populations in tumor-infiltrated immune cells compared to normal renal tissue. We further demonstrate the divergent cell transcriptional states for tumor-infiltrating CD8+ T cells and identify a MKI67 + proliferative subpopulation being a potential culprit for the progression of ccRCC. Using the SCRS gene expression, we found preferential prediction of clinical outcomes and pathological diseases by subcluster assignment. With further characterization and functional validation, our findings may reveal certain subpopulations of immune cells amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Medical Science Training Program, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Ajaykumar Vishwakarma
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Andrew P Voigt
- Medical Science Training Program, University of Iowa, Iowa City, IA, USA
| | - Andrew Bellizzi
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Jacob Kaplan
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Kenneth Nepple
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Aliasger K Salem
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Russell W Jenkins
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| | - Yousef Zakharia
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| | - Weizhou Zhang
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
89
|
Hu-Lieskovan S, Malouf GG, Jacobs I, Chou J, Liu L, Johnson ML. Addressing resistance to immune checkpoint inhibitor therapy: an urgent unmet need. Future Oncol 2021; 17:1401-1439. [PMID: 33475012 DOI: 10.2217/fon-2020-0967] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of various cancers by reversing the immunosuppressive mechanisms employed by tumors to restore anticancer immunity. Although ICIs have demonstrated substantial clinical efficacy, patient response can vary in depth and duration, and many do not respond at all or eventually develop resistance. ICI resistance mechanisms can be tumor-intrinsic, related to the tumor microenvironment or patient-specific factors. Multiple resistance mechanisms may be present within one tumor subtype, or heterogeneity exists among patients with the same tumor type. Consequently, designing effective combination treatment strategies is challenging. This review will discuss ICI resistance mechanisms, and summarize findings from key preclinical and clinical trials of ICIs, to identify potential treatment strategies or pathways to overcome ICI resistance.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Department of Medicine, Division of Oncology, Huntsman Cancer Institute / University of Utah, Salt Lake City, UT 84112, USA
| | - Gabriel G Malouf
- Department of Medical Oncology, Institut de Cancérologie de Strasbourg & Department of Functional Genomics & Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, Illkirch Cedex, Strasbourg, France
| | | | | | - Li Liu
- Pfizer Inc, San Diego, CA 92121, USA
| | - Melissa L Johnson
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN 37203, USA
| |
Collapse
|
90
|
Dobbin SJ, Petrie MC, Myles RC, Touyz RM, Lang NN. Cardiotoxic effects of angiogenesis inhibitors. Clin Sci (Lond) 2021; 135:71-100. [PMID: 33404052 PMCID: PMC7812690 DOI: 10.1042/cs20200305] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
The development of new therapies for cancer has led to dramatic improvements in survivorship. Angiogenesis inhibitors represent one such advancement, revolutionising treatment for a wide range of malignancies. However, these drugs are associated with cardiovascular toxicities which can impact optimal cancer treatment in the short-term and may lead to increased morbidity and mortality in the longer term. Vascular endothelial growth factor inhibitors (VEGFIs) are associated with hypertension, left ventricular systolic dysfunction (LVSD) and heart failure as well as arterial and venous thromboembolism, QTc interval prolongation and arrhythmia. The mechanisms behind the development of VEGFI-associated LVSD and heart failure likely involve the combination of a number of myocardial insults. These include direct myocardial effects, as well as secondary toxicity via coronary or peripheral vascular damage. Cardiac toxicity may result from the 'on-target' effects of VEGF inhibition or 'off-target' effects resulting from inhibition of other tyrosine kinases. Similar mechanisms may be involved in the development of VEGFI-associated right ventricular (RV) dysfunction. Some VEGFIs can be associated with QTc interval prolongation and an increased risk of ventricular and atrial arrhythmia. Further pre-clinical and clinical studies and trials are needed to better understand the impact of VEGFI on the cardiovascular system. Once mechanisms are elucidated, therapies can be investigated in clinical trials and surveillance strategies for identifying VEGFI-associated cardiovascular complications can be developed.
Collapse
Affiliation(s)
- Stephen J.H. Dobbin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Mark C. Petrie
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rachel C. Myles
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rhian M. Touyz
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Ninian N. Lang
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| |
Collapse
|
91
|
Xie Y, Chen Z, Zhong Q, Chen Y, Shangguan W, Xie W. Efficacy and safety of immunological checkpoint inhibitors combined with anti-angiogenic drugs in first-line treatment of metastatic renal cell carcinoma: a systematic review and meta-analysis. Transl Androl Urol 2021; 10:300-309. [PMID: 33532319 PMCID: PMC7844504 DOI: 10.21037/tau-20-969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have shown promising results for the second-line treatment of metastatic renal cell carcinoma (mRCC). Several randomized controlled trials have so far also evaluated the efficacy of ICIs for first-line treatment of mRCC. In this study, we conducted a meta-analysis of relevant studies to further clarify the efficacy and safety of ICIs combined with anti-angiogenic drugs for the treatment of mRCC. Methods We searched the PubMed, Embase, and Cochrane libraries for RCT trials of ICIs combined with anti-angiogenic drugs for first-line treatment of mRCC published before November 20, 2019. A meta-analysis was conducted based on methodological recommendations by the Cochrane Collaboration. Results Four articles with a total of 2,967 patients met the inclusion criteria. Our meta-analysis revealed that progression-free survival (PFS) and objective response rate (ORR) were significantly improved in the experimental group while there was no significant difference in overall survival (OS) (HR 0.75, 95% CI: 0.67-0.84; HR 1.43, 95% CI: 1.07-1.91; HR 0.74, 95% CI: 0.53-1.03). After stratification for PD-L1 expression, OS, PFS, and ORR of PD-L1 positive patients were significantly increased in the experimental group (HR 0.74, 95% CI: 0.56-0.96; HR 1.66, 95% CI: 1.11-2.49; HR 0.65, 95% CI: 0.57-0.75). Conclusions Immunological checkpoint inhibitors combined with anti-angiogenic drugs as a first-line treatment for mRCC improve PFS and ORR. This effect is more pronounced in PD-L1 positive patients, where ICIs also improve OS. ICIs do not increase the incidence of adverse events.
Collapse
Affiliation(s)
- Yingwei Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhiliang Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiyu Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuqing Chen
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wentai Shangguan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenlian Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
92
|
Meza L, Bergerot P, Agarwal N, Pal S. Is there a role for novel TKI/ICI combinations in metastatic renal cell carcinoma? Definitely maybe. Ann Oncol 2021; 32:12-14. [DOI: 10.1016/j.annonc.2020.10.481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 10/23/2022] Open
|
93
|
Behravesh S, Shomali N, Danbaran GR, Aslani S, Hemmatzadeh M, Hosseinzadeh R, Gowhari-Shabgah A, Mohammadi H. Cardiotoxicity of immune checkpoint inhibitors: An updated review. Biotechnol Appl Biochem 2020; 69:61-69. [PMID: 33289168 DOI: 10.1002/bab.2081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022]
Abstract
The immune checkpoint molecules are involved in the regulation of T cells in order to prevent them from attacking to sell tissues and play a role in the immune response homeostasis. Application of the immune checkpoint inhibitors (ICIs) has provided a promising therapeutic approach in pathologies where the immune system is suppressed. The extended utilization of ICIs in several cancers has caused immune-related side effects in the cardiovascular system like cardiomyopathy and myocarditis. Cardiac toxicity, one of the main side effects of the ICIs based therapeutic approach has less been concerned; however, during the last years, many cases of fatal heart failure and myocarditis have been reported in patients treated with ICIs. In this review article, we attempted to discuss the cardiac adverse effects of inhibiting different immune checkpoint molecules. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Soheil Behravesh
- Student Research Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
94
|
Nso N, Antwi-Amoabeng D, Beutler BD, Ulanja MB, Ghuman J, Hanfy A, Nimo-Boampong J, Atanga S, Doshi R, Enoru S, Gullapalli N. Cardiac adverse events of immune checkpoint inhibitors in oncology patients: A systematic review and meta-analysis. World J Cardiol 2020; 12:584-598. [PMID: 33312443 PMCID: PMC7701899 DOI: 10.4330/wjc.v12.i11.584] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are novel therapeutic agents used for various types of cancer. ICIs have revolutionized cancer treatment and improved clinical outcomes among cancer patients. However, immune-related adverse effects of ICI therapy are common. Cardiovascular immune-related adverse events (irAEs) are rare but potentially life-threatening complications.
AIM To estimate the incidence of cardiovascular irAEs among patients undergoing ICI therapy for various malignancies.
METHODS We conducted this systematic review and meta-analysis by searching PubMed, Cochrane CENTRAL, Web of Science, and SCOPUS databases for relevant interventional trials reporting cardiovascular irAEs. We performed a single-arm meta-analysis using OpenMeta [Analyst] software of the following outcomes: Myocarditis, pericardial effusion, heart failure, cardiomyopathy, atrial fibrillation, myocardial infarction, and cardiac arrest. We assessed the heterogeneity using the I2 test and managed to solve it with Cochrane’s leave-one-out method. The risk of bias was performed with the Cochrane’s risk of bias tool.
RESULTS A total of 26 studies were included. The incidence of irAEs follows: Myocarditis: 0.5% [95% confidence interval (CI): 0.1%-0.9%]; Pericardial effusion: 0.5% (95%CI: 0.1%-1.0%); Heart failure: 0.3% (95%CI: 0.0%-0.5%); Cardiomyopathy: 0.3% (95%CI: -0.1%-0.6%); atrial fibrillation: 4.6% (95%CI: 1.0%-14.1%); Myocardial infarction: 0.4% (95%CI: 0.0%-0.7%); and Cardiac arrest: 0.4% (95%CI: 0.1%-0.8%).
CONCLUSION The most common cardiovascular irAEs were atrial fibrillation, myocarditis, and pericardial effusion. Although rare, data from post market surveillance will provide estimates of the long-term prevalence and prognosis in patients with ICI-associated cardiovascular complications.
Collapse
Affiliation(s)
- Nso Nso
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens, NY 10029, United States
| | - Daniel Antwi-Amoabeng
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Bryce D Beutler
- Department of Radiology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, United States
| | - Mark B Ulanja
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Jasmine Ghuman
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Ahmed Hanfy
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Joyce Nimo-Boampong
- Department of Internal Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Sirri Atanga
- Department of Medicine, United Health Services Wilson Medical Center, Johnson City, NY 13790, United States
| | - Rajkumar Doshi
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Sostanie Enoru
- Department of Cardiovascular Disease, SUNY Downstate Health Science University, Brooklyn, NY 11203, United States
| | - Nageshwara Gullapalli
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| |
Collapse
|
95
|
Wood CG, Ferguson JE, Parker JS, Moore DT, Whisenant JG, Maygarden SJ, Wallen EM, Kim WY, Milowsky MI, Beckermann KE, Davis NB, Haake SM, Karam JA, Bortone DS, Vincent BG, Powles T, Rathmell WK. Neoadjuvant pazopanib and molecular analysis of tissue response in renal cell carcinoma. JCI Insight 2020; 5:132852. [PMID: 33208553 PMCID: PMC7710285 DOI: 10.1172/jci.insight.132852] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/08/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUNDSurgery remains the frontline therapy for patients with localized clear cell renal cell carcinoma (ccRCC); however, 20%-40% recur. Angiogenesis inhibitors have improved survival in metastatic patients and may result in responses in the neoadjuvant setting. The impact of these agents on the tumor genetic heterogeneity or the immune milieu is largely unknown. This phase II study was designed to evaluate safety, response, and effect on tumor tissue of neoadjuvant pazopanib.METHODSccRCC patients with localized disease received pazopanib (800 mg daily; median 8 weeks), followed by nephrectomy. Five tumors were examined for mutations by whole exome sequencing from samples collected before therapy and at nephrectomy. These samples underwent RNA sequencing; 17 samples were available for posttreatment assessment.RESULTSTwenty-one patients were enrolled. The overall response rate was 8 of 21 (38%). No patients with progressive disease. At 1-year, response-free survival and overall survival was 83% and 89%, respectively. The most frequent grade 3 toxicity was hypertension (33%, 7 of 21). Sequencing revealed strong concordance between pre- and posttreatment samples within individual tumors, suggesting tumors harbor stable core profiles. However, a reduction in private mutations followed treatment, suggesting a selective process favoring enrichment of driver mutations.CONCLUSIONNeoadjuvant pazopanib is safe and active in ccRCC. Future genomic analyses may enable the segregation of driver and passenger mutations. Furthermore, tumor infiltrating immune cells persist during therapy, suggesting that pazopanib can be combined with immune checkpoint inhibitors without dampening the immune response.FUNDINGSupport was provided by Novartis and GlaxoSmithKline as part of an investigator-initiated study.
Collapse
Affiliation(s)
| | - James E. Ferguson
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Dominic T. Moore
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Jennifer G. Whisenant
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Susan J. Maygarden
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Pathology
| | - Eric M. Wallen
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Oncology, and
| | - Mathew I. Milowsky
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Oncology, and
| | - Kathryn E. Beckermann
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nancy B. Davis
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Scott M. Haake
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jose A. Karam
- Department of Urology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Hematology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - W. Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
96
|
Duru G, van Egmond M, Heemskerk N. A Window of Opportunity: Targeting Cancer Endothelium to Enhance Immunotherapy. Front Immunol 2020; 11:584723. [PMID: 33262763 PMCID: PMC7686513 DOI: 10.3389/fimmu.2020.584723] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular abnormalities in tumors have a major impact on the immune microenvironment in tumors. The consequences of abnormal vasculature include increased hypoxia, acidosis, high intra-tumoral fluid pressure, and angiogenesis. This introduces an immunosuppressive microenvironment that alters immune cell maturation, activation, and trafficking, which supports tumor immune evasion and dissemination of tumor cells. Increasing data suggests that cancer endothelium is a major barrier for traveling leukocytes, ranging from a partial blockade resulting in a selective endothelial barrier, to a complete immune infiltration blockade associated with immune exclusion and immune desert cancer phenotypes. Failed immune cell trafficking as well as immunosuppression within the tumor microenvironment limits the efficacy of immunotherapeutic approaches. As such, targeting proteins with key roles in angiogenesis may potentially reduce immunosuppression and might restore infiltration of anti-tumor immune cells, creating a therapeutic window for successful immunotherapy. In this review, we provide a comprehensive overview of established as well as more controversial endothelial pathways that govern selective immune cell trafficking across cancer endothelium. Additionally, we discuss recent insights and strategies that target tumor vasculature in order to increase infiltration of cytotoxic immune cells during the therapeutic window of vascular normalization hereby improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Gizem Duru
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Niels Heemskerk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| |
Collapse
|
97
|
Hack SP, Zhu AX, Wang Y. Augmenting Anticancer Immunity Through Combined Targeting of Angiogenic and PD-1/PD-L1 Pathways: Challenges and Opportunities. Front Immunol 2020; 11:598877. [PMID: 33250900 PMCID: PMC7674951 DOI: 10.3389/fimmu.2020.598877] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy (CIT) with antibodies targeting the programmed cell death 1 protein (PD-1)/programmed cell death 1 ligand 1 (PD-L1) axis have changed the standard of care in multiple cancers. However, durable antitumor responses have been observed in only a minority of patients, indicating the presence of other inhibitory mechanisms that act to restrain anticancer immunity. Therefore, new therapeutic strategies targeted against other immune suppressive mechanisms are needed to enhance anticancer immunity and maximize the clinical benefit of CIT in patients who are resistant to immune checkpoint inhibition. Preclinical and clinical studies have identified abnormalities in the tumor microenvironment (TME) that can negatively impact the efficacy of PD-1/PD-L1 blockade. Angiogenic factors such as vascular endothelial growth factor (VEGF) drive immunosuppression in the TME by inducing vascular abnormalities, suppressing antigen presentation and immune effector cells, or augmenting the immune suppressive activity of regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages. In turn, immunosuppressive cells can drive angiogenesis, thereby creating a vicious cycle of suppressed antitumor immunity. VEGF-mediated immune suppression in the TME and its negative impact on the efficacy of CIT provide a therapeutic rationale to combine PD-1/PD-L1 antibodies with anti-VEGF drugs in order to normalize the TME. A multitude of clinical trials have been initiated to evaluate combinations of a PD-1/PD-L1 antibody with an anti-VEGF in a variety of cancers. Recently, the positive results from five Phase III studies in non-small cell lung cancer (adenocarcinoma), renal cell carcinoma, and hepatocellular carcinoma have shown that combinations of PD-1/PD-L1 antibodies and anti-VEGF agents significantly improved clinical outcomes compared with respective standards of care. Such combinations have been approved by health authorities and are now standard treatment options for renal cell carcinoma, non-small cell lung cancer, and hepatocellular carcinoma. A plethora of other randomized studies of similar combinations are currently ongoing. Here, we discuss the principle mechanisms of VEGF-mediated immunosuppression studied in preclinical models or as part of translational clinical studies. We also discuss data from recently reported randomized clinical trials. Finally, we discuss how these concepts and approaches can be further incorporated into clinical practice to improve immunotherapy outcomes for patients with cancer.
Collapse
Affiliation(s)
- Stephen P. Hack
- Product Development (Oncology), Genentech, Inc., South San Francisco, CA, United States
| | - Andrew X. Zhu
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States
- Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | - Yulei Wang
- Product Development (Oncology), Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
98
|
Ntellas P, Mavroeidis L, Gkoura S, Gazouli I, Amylidi AL, Papadaki A, Zarkavelis G, Mauri D, Karpathiou G, Kolettas E, Batistatou A, Pentheroudakis G. Old Player-New Tricks: Non Angiogenic Effects of the VEGF/VEGFR Pathway in Cancer. Cancers (Basel) 2020; 12:E3145. [PMID: 33121034 PMCID: PMC7692709 DOI: 10.3390/cancers12113145] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis has long been considered to facilitate and sustain cancer growth, making the introduction of anti-angiogenic agents that disrupt the vascular endothelial growth factor/receptor (VEGF/VEGFR) pathway an important milestone at the beginning of the 21st century. Originally research on VEGF signaling focused on its survival and mitogenic effects towards endothelial cells, with moderate so far success of anti-angiogenic therapy. However, VEGF can have multiple effects on additional cell types including immune and tumor cells, by directly influencing and promoting tumor cell survival, proliferation and invasion and contributing to an immunosuppressive microenvironment. In this review, we summarize the effects of the VEGF/VEGFR pathway on non-endothelial cells and the resulting implications of anti-angiogenic agents that include direct inhibition of tumor cell growth and immunostimulatory functions. Finally, we present how previously unappreciated studies on VEGF biology, that have demonstrated immunomodulatory properties and tumor regression by disrupting the VEGF/VEGFR pathway, now provide the scientific basis for new combinational treatments of immunotherapy with anti-angiogenic agents.
Collapse
Affiliation(s)
- Panagiotis Ntellas
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Leonidas Mavroeidis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Stefania Gkoura
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Ioanna Gazouli
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Anna-Lea Amylidi
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Alexandra Papadaki
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Davide Mauri
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Georgia Karpathiou
- Department of Pathology, University Hospital of St-Etienne, 42055 Saint Etienne, France;
| | - Evangelos Kolettas
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Biomedical Research Division, Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology, 45115 Ioannina, Greece
| | - Anna Batistatou
- Department of Pathology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - George Pentheroudakis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| |
Collapse
|
99
|
Gutierrez C, Rajendram P, Pastores SM. Toxicities Associated with Immunotherapy and Approach to Cardiotoxicity with Novel Cancer Therapies. Crit Care Clin 2020; 37:47-67. [PMID: 33190775 DOI: 10.1016/j.ccc.2020.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In recent years, major advances in oncology especially the advent of targeted agents and immunotherapies (immune checkpoint inhibitors [ICIs] and chimeric antigen receptor [CAR] T-cell therapy) have led to improved quality of life and survival rates in patients with cancer. This article focuses on the clinical features, and grading and management of toxicities associated with ICIs and CAR T-cell therapy. In addition, because cardiotoxicity is one of the most harmful effects of anticancer therapeutics, we describe the risk factors and mechanisms of cardiovascular injury associated with newer agents, screening technologies for at-risk patients, and preventive and treatment strategies.
Collapse
Affiliation(s)
- Cristina Gutierrez
- Department of Critical Care Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Prabalini Rajendram
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Stephen M Pastores
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue C-1179, New York, NY 10065, USA.
| |
Collapse
|
100
|
Cerbone L, Cattrini C, Vallome G, Latocca MM, Boccardo F, Zanardi E. Combination therapy in metastatic renal cell carcinoma: Back to the future? Semin Oncol 2020; 47:361-366. [PMID: 33168323 DOI: 10.1053/j.seminoncol.2020.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 01/12/2023]
Abstract
The treatment landscape of metastatic renal cell carcinoma (mRCC), a chemotherapy-resistant disease, has dramatically changed in the last decade after the introduction of small molecule inhibitors targeting the vascular endothelial growth factor receptor and mammalian target of rapamycin kinases. The CheckMate 025 phase III trial in second line mRCC also introduced immunotherapy with immune-checkpoint inhibitors as an option in the management of mRCC. Both small molecules and immunotherapy are used as single agents and they are associated with different toxicities. Recent data demonstrated that the combination of 2 immunotherapies, nivolumab and ipilimumab, is more effective than tyrosine kinase inhibitors (TKI) monotherapy as first line treatment in intermediate and poor risk mRCC. Furthermore, combination of immunotherapies and TKI has been tested in several trials to evaluate if the combo with agents presenting a different mechanism of action is more effective than monotherapy with TKI. During the past several years, combined therapy of cytokines doublets or cytokines and bevacizumab doublets demonstrated little improvement in clinical outcomes and a relevant toxicity profile. Conversely, the combination of new agents has been recently shown to improve survival in patients with metastatic disease, thus changing the treatment landscape of mRCC. This comprehensive review aims at summarizing the recent advances in the treatment of mRCC.
Collapse
Affiliation(s)
- Luigi Cerbone
- Dipartimento di Medicina Interna e Specialità Mediche, DIMI, Università degli Studi di Genova, Genoa, Italy; Academic Unit of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Carlo Cattrini
- Dipartimento di Medicina Interna e Specialità Mediche, DIMI, Università degli Studi di Genova, Genoa, Italy; Academic Unit of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Giacomo Vallome
- Dipartimento di Medicina Interna e Specialità Mediche, DIMI, Università degli Studi di Genova, Genoa, Italy; Academic Unit of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Maddalena Latocca
- Dipartimento di Medicina Interna e Specialità Mediche, DIMI, Università degli Studi di Genova, Genoa, Italy; Academic Unit of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Boccardo
- Dipartimento di Medicina Interna e Specialità Mediche, DIMI, Università degli Studi di Genova, Genoa, Italy; Academic Unit of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Elisa Zanardi
- Dipartimento di Medicina Interna e Specialità Mediche, DIMI, Università degli Studi di Genova, Genoa, Italy; Academic Unit of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|