51
|
Li S, Xu Q, Dai X, Zhang X, Huang M, Huang K, Shi D, Wang J, Liu L. Neoadjuvant Therapy with Immune Checkpoint Inhibitors in Gastric Cancer: A Systematic Review and Meta-Analysis. Ann Surg Oncol 2023; 30:3594-3602. [PMID: 36795255 DOI: 10.1245/s10434-023-13143-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/12/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown great promise in treating late-stage gastric cancer, but their efficacy in the neoadjuvant setting has not been studied in large cohorts. Here, we explored the efficacy and safety of neoadjuvant ICI-based therapy in locally advanced gastric cancer. PATIENTS AND METHODS We included studies containing patients with locally advanced gastric/gastroesophageal cancer who received ICI-based neoadjuvant therapy. We searched PubMed, Embase, Cochrane library, and abstracts from major international oncology conferences. We performed this meta-analysis using the META package in R.3.6.1. RESULTS Twenty-one prospective phase I/II studies comprising 687 patients were identified. The pathological complete response (pCR) rate was 0.21 (95% CI 0.18-0.24), major pathological response (MPR) rate was 0.41 (95% CI 0.31-0.52), and R0 resection rate was 0.94 (95% CI 0.92-0.96). The efficacy was highest with ICI plus radiochemotherapy, lowest with ICI alone, and in the middle with ICI and chemotherapy ± anti-angiogenesis. dMMR/MSI-H and PD-L1-high patients benefited more than pMMR/MSS and PD-L1-low patients. Grade 3 or higher toxicity rate was 0.23 (95% CI 0.13-0.38). These results exceeded those in trials of neoadjuvant chemotherapy, where the rate of pCR was 0.08 (95% CI 0.06-0.11), MPR was 0.22 (95% CI 0.19-0.26), R0 section was 0.84 (95% CI 0.80-0.87), and overall grade 3 or higher toxicity was 0.28 (95% CI 0.13-0.47) in 4800 patients across 21 studies. CONCLUSIONS In summary, the integrated results show promising efficacy and safety of ICI-based neoadjuvant therapy for locally advanced gastric cancer and support further investigation in large multicenter randomized trials.
Collapse
Affiliation(s)
- Song Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qian Xu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xin Dai
- Department of Medical Oncology, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xue Zhang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Miao Huang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Kai Huang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Duanbo Shi
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jian Wang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lian Liu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
52
|
Baudoux N, Friedlaender A, Addeo A. Evolving Therapeutic Scenario of Stage III Non-Small-Cell Lung Cancer. Clin Med Insights Oncol 2023; 17:11795549231152948. [PMID: 36818454 PMCID: PMC9932776 DOI: 10.1177/11795549231152948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
Lung cancer remains the leading cause of cancer-related death with an incidence that continues to increase in both sexes and all ages. However, 80% to 90% of lung cancers are non-small cell lung cancer (NSCLC) and the remaining 10% to 20% are small cell lung cancer. Adenocarcinoma is the most common histologic subtype of lung cancer worldwide. More frequently, lung cancer diagnosis is made in advanced stages. Stage III NSCLC refers to locoregionally advanced disease without metastases and represents about 30% NSCLC cases. Despite the absence of metastases at diagnosis, the outcome is generally poor. Stage III comprises a heterogeneous group and optimal management requires the input of a multidisciplinary team. All modalities of oncologic treatment are involved: surgery, chemotherapy, radiotherapy, and more recently, immunotherapy and targeted therapy. We will discuss the different therapeutic options in stage III NSCLC, both in operable and inoperable scenarios, and the role of immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Nathalie Baudoux
- Oncology Department, Geneva University
Hospitals, Geneva, Switzerland
| | - Alex Friedlaender
- Oncology Department, Geneva University
Hospitals, Geneva, Switzerland
- Oncology Service, Clinique Générale
Beaulieu, Geneva, Switzerland
| | - Alfredo Addeo
- Oncology Department, Geneva University
Hospitals, Geneva, Switzerland
- Alfredo Addeo, Oncology Department, Geneva
University Hospitals, Geneva, 1205, Switzerland.
| |
Collapse
|
53
|
He T, Hu M, Zhu S, Shen M, Kou X, Liang X, Li L, Li X, Zhang M, Wu Q, Gong C. A tactical nanomissile mobilizing antitumor immunity enables neoadjuvant chemo-immunotherapy to minimize postsurgical tumor metastasis and recurrence. Acta Pharm Sin B 2023; 13:804-818. [PMID: 36873172 PMCID: PMC9979264 DOI: 10.1016/j.apsb.2022.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/07/2022] [Accepted: 06/19/2022] [Indexed: 11/27/2022] Open
Abstract
Neoadjuvant chemotherapy has become an indispensable weapon against high-risk resectable cancers, which benefits from tumor downstaging. However, the utility of chemotherapeutics alone as a neoadjuvant agent is incapable of generating durable therapeutic benefits to prevent postsurgical tumor metastasis and recurrence. Herein, a tactical nanomissile (TALE), equipped with a guidance system (PD-L1 monoclonal antibody), ammunition (mitoxantrone, Mit), and projectile bodies (tertiary amines modified azobenzene derivatives), is designed as a neoadjuvant chemo-immunotherapy setting, which aims at targeting tumor cells, and fast-releasing Mit owing to the intracellular azoreductase, thereby inducing immunogenic tumor cells death, and forming an in situ tumor vaccine containing damage-associated molecular patterns and multiple tumor antigen epitopes to mobilize the immune system. The formed in situ tumor vaccine can recruit and activate antigen-presenting cells, and ultimately increase the infiltration of CD8+ T cells while reversing the immunosuppression microenvironment. Moreover, this approach provokes a robust systemic immune response and immunological memory, as evidenced by preventing 83.3% of mice from postsurgical metastasis or recurrence in the B16-F10 tumor mouse model. Collectively, our results highlight the potential of TALE as a neoadjuvant chemo-immunotherapy paradigm that can not only debulk tumors but generate a long-term immunosurveillance to maximize the durable benefits of neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Tao He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingxing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shunyao Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meiling Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaorong Kou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiuqi Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinchao Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Miaomiao Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qinjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
54
|
Lv H, Huang C, Li J, Zhang F, Gai C, Liu Z, Xu S, Wang M, Li Z, Tian Z. The survival outcomes of neoadjuvant sintilimab combined with chemotherapy in patients with locally advanced esophageal squamous cell carcinoma. Front Immunol 2023; 13:1100750. [PMID: 36741358 PMCID: PMC9892710 DOI: 10.3389/fimmu.2022.1100750] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
Background Neoadjuvant programmed death receptor-1 (PD-1) inhibitor combined with chemotherapy has been reported to improve the pathological response of locally advanced esophageal squamous cell carcinoma (ESCC), but the systematic report on survival follow-up is quite few. This study we will report the survival follow-up outcomes after a median follow-up of 21.1 months. Methods This was a real-world retrospective study. Locally advanced ESCC patients treated with neoadjuvant sintilimab combined with albumin-bound paclitaxel and nedaplatin followed by surgery and completed at least 1-year follow-up were reviewed. The primary outcome was disease-free survival (DFS) at 24 months. The secondary outcome was overall survival (OS) at 24 months. Results Ninety eligible patients were included in the analysis between July 2019 and October 2021. The median number of neoadjuvant cycles was 3 (range 2-4). All patients achieved R0 resection. With a median follow-up of 21.1 months (range 14.0-39.0), the median DFS and median OS had not reached, 2-year DFS rate was 78.3% (95%CI 68.8%-89.1%) and 2-years OS rate was 88.0% (95%CI 80.6%-96.0%). Postoperative pathological stage, pCR, MPR, tumor down-staging were significantly correlated with favorable survival outcome. Univariable and multivariable Cox regression analysis identified cycle number of neoadjuvant treatment as independent predictor of DFS. Conclusion Our results preliminarily show a survival benefit of neoadjuvant sintilimab combined with chemotherapy in locally advanced ESCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
55
|
Versluis JM, Menzies AM, Sikorska K, Rozeman EA, Saw RPM, van Houdt WJ, Eriksson H, Klop WMC, Ch'ng S, van Thienen JV, Mallo H, Gonzalez M, Torres Acosta A, Grijpink-Ongering LG, van der Wal A, Bruining A, van de Wiel BA, Scolyer RA, Haanen JBAG, Schumacher TN, van Akkooi ACJ, Long GV, Blank CU. Survival update of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma in the OpACIN and OpACIN-neo trials. Ann Oncol 2023; 34:420-430. [PMID: 36681299 DOI: 10.1016/j.annonc.2023.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Neoadjuvant ipilimumab plus nivolumab has yielded high response rates in patients with macroscopic stage III melanoma. These response rates translated to high short-term survival rates. However, data on long-term survival and disease recurrence are lacking. PATIENTS AND METHODS In OpACIN, 20 patients with macroscopic stage III melanoma were randomized to ipilimumab 3 mg/kg plus nivolumab 1 mg/kg q3w four cycles of adjuvant or split two cycles of neoadjuvant and two adjuvant. In OpACIN-neo, 86 patients with macroscopic stage III melanoma were randomized to arm A (2× ipilimumab 3 mg/kg plus nivolumab 1 mg/kg q3w; n = 30), arm B (2× ipilimumab 1 mg/kg plus nivolumab 3 mg/kg q3w; n = 30), or arm C (2× ipilimumab 3 mg/kg q3w plus 2× nivolumab 3 mg/kg q2w; n = 26) followed by surgery. RESULTS The median recurrence-free survival (RFS) and overall survival (OS) were not reached in either trial. After a median follow-up of 69 months for OpACIN, 1/7 patients with a pathologic response to neoadjuvant therapy had disease recurrence. The estimated 5-year RFS and OS rates for the neoadjuvant arm were 70% and 90% versus 60% and 70% for the adjuvant arm. After a median follow-up of 47 months for OpACIN-neo, the estimated 3-year RFS and OS rates were 82% and 92%, respectively. The estimated 3-year RFS rate for OpACIN-neo was 95% for patients with a pathologic response versus 37% for patients without a pathologic response (P < 0.001). In multiple regression analyses, pathologic response was the strongest predictor of disease recurrence. Of the 12 patients with distant disease recurrence after neoadjuvant therapy, 5 responded to subsequent anti-PD-1 and 8 to targeted therapy, although 7 patients showed progression after the initial response. CONCLUSIONS Updated data confirm the high survival rates after neoadjuvant combination checkpoint inhibition in macroscopic stage III melanoma, especially for patients with a pathologic response. Pathologic response is the strongest surrogate marker for long-term outcome.
Collapse
Affiliation(s)
- J M Versluis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - K Sikorska
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - E A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Department of Surgery, Mater Hospital, Sydney; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - W J van Houdt
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - H Eriksson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm; Department of Oncology/Skin Cancer Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - W M C Klop
- Departments of, Head and Neck Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney; Department of Surgery, Mater Hospital, Sydney; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - J V van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - H Mallo
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Gonzalez
- Melanoma Institute Australia, The University of Sydney, Sydney
| | - A Torres Acosta
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - A van der Wal
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Bruining
- Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - B A van de Wiel
- Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - J B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - T N Schumacher
- Department of Hematology, Leiden University Medical Center, Leiden; Division of Molecular Oncology and Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam
| | - A C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - G V Long
- Melanoma Institute Australia, The University of Sydney, Sydney; Faculty of Medicine and Health, The University of Sydney, Sydney; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - C U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam; Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
56
|
Gorry C, McCullagh L, O'Donnell H, Barrett S, Schmitz S, Barry M, Curtin K, Beausang E, Barry R, Coyne I. Neoadjuvant treatment for stage III and IV cutaneous melanoma. Cochrane Database Syst Rev 2023; 1:CD012974. [PMID: 36648215 PMCID: PMC9844053 DOI: 10.1002/14651858.cd012974.pub2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cutaneous melanoma is amongst the most aggressive of all skin cancers. Neoadjuvant treatment is a form of induction therapy, given to shrink a cancerous tumour prior to the main treatment (usually surgery). The purpose is to improve survival and surgical outcomes. This review systematically appraises the literature investigating the use of neoadjuvant treatment for stage III and IV cutaneous melanoma. OBJECTIVES To assess the effects of neoadjuvant treatment in adults with stage III or stage IV melanoma according to the seventh edition American Joint Committee on Cancer (AJCC) staging system. SEARCH METHODS We searched the following databases up to 10 August 2021 inclusive: Cochrane Skin Specialised Register, CENTRAL, MEDLINE, Embase, LILACS and four trials registers, together with reference checking and contact with study authors to identify additional studies. We also handsearched proceedings from specific conferences from 2016 to 2020 inclusive. SELECTION CRITERIA Randomised controlled trials (RCTs) of people with stage III and IV melanoma, comparing neoadjuvant treatment strategies (using targeted treatments, immunotherapies, radiotherapy, topical treatments or chemotherapy) with any of these agents or current standard of care (SOC), were eligible for inclusion. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Primary outcomes were overall survival (OS) and adverse effects (AEs). Secondary outcomes included time to recurrence (TTR), quality of life (QOL), and overall response rate (ORR). We used GRADE to evaluate the certainty of the evidence. MAIN RESULTS We included eight RCTs involving 402 participants. Studies enrolled adults, mostly with stage III melanoma, investigated immunotherapies, chemotherapy, or targeted treatments, and compared these with surgical excision with or without adjuvant treatment. Duration of follow-up and therapeutic regimens varied, which, combined with heterogeneity in the population and definitions of the endpoints, precluded meta-analysis of all identified studies. We performed a meta-analysis including three studies. We are very uncertain if neoadjuvant treatment increases OS when compared to no neoadjuvant treatment (hazard ratio (HR) 0.43, 95% confidence interval (CI) 0.15 to 1.21; 2 studies, 171 participants; very low-certainty evidence). Neoadjuvant treatment may increase the rate of AEs, but the evidence is very uncertain (26% versus 16%, risk ratio (RR) 1.58, 95% CI 0.97 to 2.55; 2 studies, 162 participants; very low-certainty evidence). We are very uncertain if neoadjuvant treatment increases TTR (HR 0.51, 95% CI 0.22 to 1.17; 2 studies, 171 participants; very low-certainty evidence). Studies did not report ORR as a comparative outcome or measure QOL data. We are very uncertain whether neoadjuvant targeted treatment with dabrafenib and trametinib increases OS (HR 0.28, 95% CI 0.03 to 2.25; 1 study, 21 participants; very low-certainty evidence) or TTR (HR 0.02, 95% CI 0.00 to 0.22; 1 study, 21 participants; very low-certainty evidence) when compared to surgery. The study did not report comparative rates of AEs and overall response, and did not measure QOL. We are very uncertain if neoadjuvant immunotherapy with talimogene laherparepvec increases OS when compared to no neoadjuvant treatment (HR 0.49, 95% CI 0.15 to 1.64; 1 study, 150 participants, very low-certainty evidence). It may have a higher rate of AEs, but the evidence is very uncertain (16.5% versus 5.8%, RR 2.84, 95% CI 0.96 to 8.37; 1 study, 142 participants; very low-certainty evidence). We are very uncertain if it increases TTR (HR 0.75, 95% CI 0.31 to 1.79; 1 study, 150 participants; very low-certainty evidence). The study did not report comparative ORRs or measure QOL. OS was not reported for neoadjuvant immunotherapy (combined ipilimumab and nivolumab) when compared to the combination of ipilimumab and nivolumab as adjuvant treatment. There may be little or no difference in the rate of AEs between these treatments (9%, RR 1.0, 95% CI 0.75 to 1.34; 1 study, 20 participants; low-certainty evidence). The study did not report comparative ORRs or measure TTR and QOL. Neoadjuvant immunotherapy (combined ipilimumab and nivolumab) likely results in little to no difference in OS when compared to neoadjuvant nivolumab monotherapy (P = 0.18; 1 study, 23 participants; moderate-certainty evidence). It may increase the rate of AEs, but the certainty of this evidence is very low (72.8% versus 8.3%, RR 8.73, 95% CI 1.29 to 59; 1 study, 23 participants); this trial was halted early due to observation of disease progression preventing surgical resection in the monotherapy arm and the high rate of treatment-related AEs in the combination arm. Neoadjuvant combination treatment may lead to higher ORR, but the evidence is very uncertain (72.8% versus 25%, RR 2.91, 95% CI 1.02 to 8.27; 1 study, 23 participants; very low-certainty evidence). It likely results in little to no difference in TTR (P = 0.19; 1 study, 23 participants; low-certainty evidence). The study did not measure QOL. OS was not reported for neoadjuvant immunotherapy (combined ipilimumab and nivolumab) when compared to neoadjuvant sequential immunotherapy (ipilimumab then nivolumab). Only Grade 3 to 4 immune-related AEs were reported; fewer were reported with combination treatment, and the sequential treatment arm closed early due to a high incidence of severe AEs. The neoadjuvant combination likely results in a higher ORR compared to sequential neoadjuvant treatment (60.1% versus 42.3%, RR 1.42, 95% CI 0.87 to 2.32; 1 study, 86 participants; low-certainty evidence). The study did not measure TTR and QOL. No data were reported on OS, AEs, TTR, or QOL for the comparison of neoadjuvant interferon (HDI) plus chemotherapy versus neoadjuvant chemotherapy. Neoadjuvant HDI plus chemotherapy may have little to no effect on ORR, but the evidence is very uncertain (33% versus 22%, RR 1.75, 95% CI 0.62 to 4.95; 1 study, 36 participants; very low-certainty evidence). AUTHORS' CONCLUSIONS We are uncertain if neoadjuvant treatment increases OS or TTR compared with no neoadjuvant treatment, and it may be associated with a slightly higher rate of AEs. There is insufficient evidence to support the use of neoadjuvant treatment in clinical practice. Priorities for research include the development of a core outcome set for neoadjuvant trials that are adequately powered, with validation of pathological and radiological responses as intermediate endpoints, to investigate the relative benefits of neoadjuvant treatment compared with adjuvant treatment with immunotherapies or targeted therapies.
Collapse
Affiliation(s)
- Claire Gorry
- National Centre for Pharmacoeconomics, St James's Hospital, Dublin, Ireland
| | - Laura McCullagh
- National Centre for Pharmacoeconomics, St James's Hospital, Dublin, Ireland
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Helen O'Donnell
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Sarah Barrett
- Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity St James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Susanne Schmitz
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Michael Barry
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Kay Curtin
- Melanoma Support Ireland, Dublin, Ireland
| | - Eamon Beausang
- Plastic and Reconstructive Surgery, St James's Hospital, Dublin, Ireland
| | - Rupert Barry
- Department of Dermatology, St James's Hospital, Dublin, Ireland
| | - Imelda Coyne
- School of Nursing & Midwifery, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
57
|
Jacoberger-Foissac C, Allard B, Allard D, Stagg J. Assessing the Efficacy of Immune Checkpoint Inhibitors in Preclinical Tumor Models. Methods Mol Biol 2023; 2614:151-169. [PMID: 36587125 DOI: 10.1007/978-1-0716-2914-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The identification of novel immune-related targets that can reactivate or enhance antitumor immunity is a very active field of cancer research. In this context, syngeneic tumor models are often used during the preclinical development of immunotherapies to assess their efficacy and analyze the immune system and tumor cell interaction. Here, we present the practical procedures to generate subcutaneous tumors and experimental lung metastases used to evaluate the antitumor activity of your immunotherapy of interest. We also describe a method to quantify contrasted lung metastasis burden by imaging. Finally, we present a protocol to perform orthotopic injection of breast tumor cells in the mammary fat pad followed by tumor resection for the study of spontaneous metastases and evaluation of neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Celia Jacoberger-Foissac
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montréal, QC, Canada
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montréal, QC, Canada
| | - David Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montréal, QC, Canada
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montréal, QC, Canada.
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
58
|
Sun W, Dai L, Cao Y, Pan P, Zhi L, Wang X, Yuan X, Gao Z, Guo S, Liu G, Yin J, Xie L, Wang L, Wang Y, Li W, Li H, Jia Y. Monocytes reprogrammed by tumor microparticle vaccine inhibit tumorigenesis and tumor development. Cancer Nanotechnol 2023; 14:34. [PMID: 37089435 PMCID: PMC10106871 DOI: 10.1186/s12645-023-00190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/06/2023] [Indexed: 04/25/2023] Open
Abstract
Tumor microparticles (T-MPs) are considered as a tumor vaccine candidate. Although some studies have analyzed the mechanism of T-MPs as tumor vaccine, we still lack understanding of how T-MPs stimulate a strong anti-tumor immune response. Here, we show that T-MPs induce macrophages to release a key chemotactic factor CCL2, which attracts monocytes to the vaccine injection site and enhances endocytosis of antigen. Monocytes subsequently enter the draining lymph node, and differentiate into monocyte-derived DCs (moDCs), which present tumor antigens to T lymphocytes and deliver a potent anti-tumor immune response. Mechanically, T-MPs activate the cGAS-STING signaling through DNA fragments, and then induce monocytes to upregulate the expression of IRF4, which is a key factor for monocyte differentiation into moDCs. More importantly, monocytes that have endocytosed T-MPs acquire the ability to treat tumors. Collectively, this work might provide novel vaccination strategy for the development of tumor vaccines and facilitate the application of T-MPs for clinic oncotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12645-023-00190-x.
Collapse
Affiliation(s)
- Weiwei Sun
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Lili Dai
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Yuqing Cao
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Pengtao Pan
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Lijuan Zhi
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Xinke Wang
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Xinzhong Yuan
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Zi Gao
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Sheng Guo
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Guoyan Liu
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Junlei Yin
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Liangliang Xie
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Liping Wang
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Yanling Wang
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Wensheng Li
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Hong Li
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| | - Yunjie Jia
- School of Medicine, Xinxiang University, Jinsui Road 191, Xinxiang, 453003 China
| |
Collapse
|
59
|
Alvarez M, Molina C, Garasa S, Ochoa MC, Rodriguez-Ruiz ME, Gomis G, Cirella A, Olivera I, Glez-Vaz J, Gonzalez-Gomariz J, luri-Rey C, azpilikueta A, Bolaños E, Teijeira A, Berraondo P, Quintero M, Melero I. Intratumoral neoadjuvant immunotherapy based on the BO-112 viral RNA mimetic. Oncoimmunology 2023; 12:2197370. [PMID: 37035637 PMCID: PMC10078127 DOI: 10.1080/2162402x.2023.2197370] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023] Open
Abstract
BO-112 is a poly I:C-based viral mimetic that exerts anti-tumor efficacy when intratumorally delivered in mouse models. Intratumoral BO-112 synergizes in mice with systemic anti-PD-1 mAbs and this combination has attained efficacy in PD1-refractory melanoma patients. We sought to evaluate the anti-tumor efficacy of BO-112 pre-surgically applied in neoadjuvant settings to mouse models. We have observed that repeated intratumoral injections of BO-112 prior to surgical excision of the primary tumor significantly reduced tumor metastasis from orthotopically implanted 4T1-derived tumors and subcutaneous MC38-derived tumors in mice. Such effects were enhanced when combined with systemic anti-PD-1 mAb. The anti-tumor efficacy of this neoadjuvant immunotherapy approach depended on the presence of antigen-specific effector CD8 T cells and cDC1 antigen-presenting cells. Since BO-112 has been successful in phase-two clinical trials for metastatic melanoma, these results provide a strong rationale for translating this pre-surgical strategy into clinical settings, especially in combination with standard-of-care checkpoint inhibitors.
Collapse
Affiliation(s)
- Maite Alvarez
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- CONTACT Maite Alvarez
| | - Carmen Molina
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Saray Garasa
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maria C. Ochoa
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Maria E Rodriguez-Ruiz
- Departments of Immunology and Oncology (CCUN), Clínica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Gomis
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Assunta Cirella
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Irene Olivera
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Javier Glez-Vaz
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Jose Gonzalez-Gomariz
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - carlos luri-Rey
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - arantza azpilikueta
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Elixabet Bolaños
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Alvaro Teijeira
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Pedro Berraondo
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Ignacio Melero
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Departments of Immunology and Oncology (CCUN), Clínica Universidad de Navarra, Pamplona, Spain
- Nuffield Department of Medicine and Oxford Center for Immuno-Oncology, University of Oxford, Oxford, UK
- Ignacio Melero Program for Immunology and Immunotherapy, CIMA Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
60
|
Viscardi G, Vitiello F, Servetto A, Gristina V, Pizzutilo EG, Canciello MA, Medusa PM, Salomone F, Di Guida G, Mollica M, Aronne L, Scaramuzzi R, Napolitano F, Battiloro C, Caputo F, Gilli M, Totaro G, Curcio C, Rocco D, Montesarchio V. Moving Immune Checkpoint Inhibitors to Early Non-Small Cell Lung Cancer: A Narrative Review. Cancers (Basel) 2022; 14:cancers14235810. [PMID: 36497292 PMCID: PMC9735901 DOI: 10.3390/cancers14235810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Since prognosis of early-stage non-small cell lung cancer (NSCLC) remains dismal for common relapses after curative surgery, considerable efforts are currently focused on bringing immunotherapy into neoadjuvant and adjuvant settings. Previously, perioperative chemotherapy showed only a modest but significative improvement in overall survival. The presence of broad tumor neoantigens load at primary tumor prior to surgery as well as the known immunosuppressive status following resection represent the main rationale for immunotherapy in early disease. Several trials have been conducted in recent years, leading to atezolizumab and nivolumab approval in the adjuvant and neoadjuvant setting, respectively, and perioperative immunotherapy in NSCLC remains a field of active clinical and preclinical investigation. Unanswered questions in perioperative therapy in NSCLC include the optimal sequence and timing of chemotherapy and immunotherapy, the potential of combination strategies, the role of predictive biomarkers for patient selection and the choice of useful endpoints in clinical investigation.
Collapse
Affiliation(s)
- Giuseppe Viscardi
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via Sergio Pansini 5, 80131 Naples, Italy
- Correspondence:
| | - Fabiana Vitiello
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Alberto Servetto
- Medical Oncology, Department of Clinical Medicine and Surgery, Università degli Studi di Napoli “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Valerio Gristina
- Medical Oncology, Department of Surgical, Oncological and Oral Sciences, Università degli Studi di Palermo, Via Liborio Giuffrè 5, 90127 Palermo, Italy
| | - Elio Gregory Pizzutilo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza dell’Ospedale Maggiore 3, 20162 Milan, Italy
- Departmento of Oncology and Hematology, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Maria Anna Canciello
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Paola Maria Medusa
- Pneumology Unit, Università degli Studi della Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Fabio Salomone
- Medical Oncology, Department of Clinical Medicine and Surgery, Università degli Studi di Napoli “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Gaetano Di Guida
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Mariano Mollica
- Respiratory Pathophysiology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Luigi Aronne
- Pneumology Unit, Università degli Studi della Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Roberto Scaramuzzi
- Thoracic Surgery, Department of General and Specialistic Surgery, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Filomena Napolitano
- Thoracic Surgery, Department of General and Specialistic Surgery, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Ciro Battiloro
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Francesca Caputo
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Marina Gilli
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Giuseppe Totaro
- Radiotherapy Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Carlo Curcio
- Thoracic Surgery, Department of General and Specialistic Surgery, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Danilo Rocco
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Vincenzo Montesarchio
- Medical Oncology, Department of Pneumology and Oncology, AORN Ospedali dei Colli, Via Leonardo Bianchi, 80131 Naples, Italy
| |
Collapse
|
61
|
Yang Y, Tan L, Hu J, Li Y, Mao Y, Tian Z, Zhang B, Ma J, Li H, Chen C, Chen K, Han Y, Chen L, Liu J, Yu B, Yu Z, Li Z. Safety and efficacy of neoadjuvant treatment with immune checkpoint inhibitors in esophageal cancer: real-world multicenter retrospective study in China. Dis Esophagus 2022; 35:6596998. [PMID: 35649396 DOI: 10.1093/dote/doac031] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/26/2022] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have shown a powerful benefit in the neoadjuvant therapy for esophageal cancer, but evidence for its safety and efficacy is limited and may not reflect real-world practice. We retrospectively reviewed the database of treatment-naive patients from 15 esophageal cancer centers in China who received ICIs as neoadjuvant treatment for locally advanced esophageal cancer from May 2019 to December 2020. The primary endpoints were rate and severity of treatment-related adverse events (TRAEs) and immune-related adverse events (irAEs). Secondary endpoints included pathologically complete response (pCR) rate, R0 resection rate, mortality and morbidity. Among the 370 patients, 311 (84.1%) were male with a median age of 63 (range: 30-81) years and stage III or IVa disease accounted for 84.1% of these patients. A total of 299 (80.8%) patients were treated with ICIs and chemotherapy. TRAEs were observed in 199 (53.8%) patients with low severity (grade 1-2, 39.2%; grade 3-4, 13.2%; grade 5, 1.4%), and irAEs occurred in 24.3% of patients and were mostly of grade 1-2 severity (21.1%). A total of 341 (92.2%) patients had received surgery and R0 resection was achieved in 333 (97.7%) patients. The local pCR rate in primary tumor was 34.6%, including 25.8% of ypT0N0 and 8.8% of ypT0N+. The rate of postoperative complications was 41.4% and grade 3 or higher complications occurred in 35 (10.3%) patients. No death was observed within 30 days after surgery, and three patients (0.9%) died within 90 days postoperatively. This study shows acceptable toxicity of neoadjuvant immunotherapy for locally advanced esophageal cancer in real-world data. Long-term survival results are pending for further investigations.
Collapse
Affiliation(s)
- Yang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai, P. R. China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Zhejiang, P. R. China
| | - Yin Li
- Department of Thoracic Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yousheng Mao
- Department of Thoracic Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Baihua Zhang
- Department of Thoracic Surgery, Hunan Cancer Hospital, Changsha, P. R. China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, P. R. China
| | - Keneng Chen
- Department of Thoracic Surgery, Beijing Cancer Hospital, Beijing, P. R. China
| | - Yongtao Han
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Chengdu, P. R. China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Junfeng Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Zhentao Yu
- Department of Thoracic Surgery, Tianjin Medical University Cancer Hospital, Tianjin, P. R. China.,Department of Thoracic Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, P. R. China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | | |
Collapse
|
62
|
Li Y, Xue C, Gao Z, Li K, Xu H, Zhu Z. Efficacy of neoadjuvant immunotherapy in advanced colorectal cancer: a meta-analysis of cross-sectional studies. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04402-6. [PMID: 36260157 DOI: 10.1007/s00432-022-04402-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/05/2022] [Indexed: 10/24/2022]
Abstract
BACKGROUND Although neoadjuvant immunotherapy is being widely studied, there is no consensus on its efficacy in microsatellite-stable (MSS) or mismatch repair proficient (pMMR) colorectal cancer (CRC). This meta-analysis aimed to evaluate studies on neoadjuvant immunotherapy for advanced CRC to assess its efficacy and provide new clinical guidelines. METHODS We searched literature databases to identify studies that assessed the efficacy of neoadjuvant immunotherapy in advanced CRC. The outcomes evaluated were pathological complete response (pCR), major pathological response (MPR), R0 resection, and anal preservation rates. Heterogeneity among the included studies was assessed by sensitivity analysis, and publication bias was evaluated using Begg and Egger tests. RESULTS Eleven articles were included in the analysis. The pCR, MPR, R0 resection, and anal preservation rates reported in these studies were 39 and 49, 97, and 76%, respectively. The MSI-H and MSS groups had pooled pCR rates of 70 and 24%, respectively. The pCR rates for the induction, consolidation, and concurrent immuno-chemoradiotherapy (CRT) subgroups were 43, 33, and 27%, respectively, and those for the single and double immunotherapy subgroups were 34 and 40%, respectively. CONCLUSION Neoadjuvant immunotherapy combined with CRT is effective in treating MSI-H/dMMR advanced CRC. It could also be a new first-line therapeutic option for MSS/pMMR advanced CRC.
Collapse
Affiliation(s)
- Yuegang Li
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China.,Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences, Langfang, 065001, China
| | - Chi Xue
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Ziming Gao
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Kai Li
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Huimian Xu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Zhi Zhu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
63
|
Ho J, Mattei J, Tetzlaff M, Williams MD, Davies MA, Diab A, Oliva ICG, McQuade J, Patel SP, Tawbi H, Wong MK, Fisher SB, Hanna E, Keung EZ, Ross M, Weiser R, Su SY, Frumovitz M, Meyer LA, Jazaeri A, Pettaway CA, Guadagnolo BA, Bishop AJ, Mitra D, Farooqi A, Bassett R, Faria S, Nagarajan P, Amaria RN. Neoadjuvant checkpoint inhibitor immunotherapy for resectable mucosal melanoma. Front Oncol 2022; 12:1001150. [PMID: 36324592 PMCID: PMC9618687 DOI: 10.3389/fonc.2022.1001150] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Background Neoadjuvant checkpoint inhibition (CPI) has recently demonstrated impressive outcomes in patients with stage 3 cutaneous melanoma. However, the safety, efficacy, and outcome of neoadjuvant CPI in patients with mucosal melanoma (MM) are not well studied as MM is a rare melanoma subtype. CPI such as combination nivolumab and ipilimumab achieves response rates of 37-43% in unresectable or metastatic MM but there is limited data regarding the efficacy of these agents in the preoperative setting. We hypothesize that neoadjuvant CPI is a safe and feasible approach for patients with resectable MM. Method Under an institutionally approved protocol, we identified adult MM patients with resectable disease who received neoadjuvant anti-PD1 +/- anti-CTLA4 between 2015 to 2019 at our institution. Clinical information include age, gender, presence of nodal involvement or satellitosis, functional status, pre-treatment LDH, tumor mutation status, and treatment data was collected. Outcomes include event free survival (EFS), overall survival (OS), objective response rate (ORR), pathologic response rate (PRR), and grade ≥3 toxicities. Results We identified 36 patients. Median age was 62; 58% were female. Seventy-eight percent of patients received anti-PD1 + anti-CTLA4. Node positive disease or satellite lesions was present at the time of treatment initiation in 47% of patients. Primary sites of disease were anorectal (53%), urogenital (25%), head and neck (17%), and esophageal (6%). A minority of patients did not undergo surgery due to complete response (n=3, 8%) and disease progression (n=6, 17%), respectively. With a median follow up of 37.9 months, the median EFS was 9.2 months with 3-year EFS rate of 29%. Median OS had not been reached and 3-year OS rate was 55%. ORR was 47% and PRR was 35%. EFS was significantly higher for patients with objective response and for patients with pathologic response. OS was significantly higher for patients with pathologic response. Grade 3 toxicities were reported in 39% of patients. Conclusion Neoadjuvant CPI for resectable MM is a feasible approach with signs of efficacy and an acceptable safety profile. As there is currently no standard approach for resectable MM, this study supports further investigations using neoadjuvant therapy for these patients.
Collapse
Affiliation(s)
- Joel Ho
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jane Mattei
- Oncology Department, Hospital Moinhos de Vento, Porto Alegre, Brazil
| | - Michael Tetzlaff
- Division of Dermatopathology, University of California San Francisco (UCSF), San Francisco, CA, United States
| | - Michelle D. Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Isabella C. Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sapna P. Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael K. Wong
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sarah B. Fisher
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ehab Hanna
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Merrick Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Roi Weiser
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shirley Y. Su
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Larissa A. Meyer
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Curtis A. Pettaway
- Department of Urologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - B. Ashleigh Guadagnolo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew J. Bishop
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Devarati Mitra
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ahsan Farooqi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Silvana Faria
- Department of Abdominal Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Priyadharsini Nagarajan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rodabe N. Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
64
|
Wu Z, Li M, Wang L, Paul A, Raman JD, Necchi A, Psutka SP, Buonerba C, Zargar H, Black PC, Derweesh IH, Mir MC, Uzzo RG, Pandolfo SD, Autorino R, DI Lorenzo G. Neoadjuvant systemic therapy in patients undergoing nephroureterectomy for urothelial cancer: a multidisciplinary systematic review and critical analysis. Minerva Urol Nephrol 2022; 74:518-527. [PMID: 35383431 DOI: 10.23736/s2724-6051.22.04659-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION The benefit of neoadjuvant systemic therapy (NAST) is not yet supported by randomized controlled trials in upper tract urothelial carcinoma (UTUC), but the evidence is increasing. This narrative systematic review was conducted to evaluate the available evidence on the role of NAST in patients undergoing radical nephroureterectomy (RNU) for UTUC. EVIDENCE ACQUISITION We searched for all relevant articles or conference abstracts published and indexed in PubMed, Embase, and Scopus on July 19, 2021. The study was reported according to the PRISMA criteria and designed within the PICOS framework. We included studies comparing patients with non-metastatic UTUC who received neoadjuvant chemotherapy (NAC) or immunotherapy (NAI) with patients who underwent definitive surgery alone or surgery plus adjuvant systemic therapy. Prospective uncontrolled studies were also included. EVIDENCE SYNTHESIS We identified 27 reports (NAC, N.=24 and NAI, N.=3) published between 2010 and 2021. Twenty of the 24 studies on NAC were retrospective comparative analyses, whereas the remaining four were prospective single-arm studies. One of the three NAI studies exclusively enrolled patients with UTUC. NAC was associated with improved survival and better pathological response relative to surgery alone, but there was no clear advantage when compared to surgery plus adjuvant chemotherapy. Overall, the drug-induced toxicity and risk of disease progression were acceptable but the inherent bias across study designs, inadequate reporting and heterogeneous definition of primary outcomes render it difficult to synthesize results, compare centers, and inform practice. CONCLUSIONS The current level of evidence supporting NAST for UTUC is relatively low and the inability to predict responsiveness and thereby pinpoint the optimal candidates remains a major challenge. There is a need to compare NAST to adjuvant therapies using clearly defined primary endpoints as minimum reporting standards developed by a multidisciplinary team.
Collapse
Affiliation(s)
- Zhenjie Wu
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mingmin Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China -
| | - Asit Paul
- Division of Hematology, Oncology and Palliative Care, Department of Internal Medicine, VCU Health, Richmond, VA, USA
| | - Jay D Raman
- Department of Urology, Penn State Health, Hershey, PA, USA
| | - Andrea Necchi
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Sarah P Psutka
- Department of Urology, Seattle Cancer Care Alliance, University of Washington, Seattle, WA, USA
| | - Carlo Buonerba
- Department of Oncology and Hematology, Regional Reference Center for Rare Tumors, University of Naples Federico II, Naples, Italy
| | - Homayoun Zargar
- Unit of Surgery, Department of Urology, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - Peter C Black
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Ithaar H Derweesh
- Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Maria C Mir
- Department of Urology, Valencian Oncology Institute Foundation, FIVO, Valencia, Spain
| | - Robert G Uzzo
- Division of Urological Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | - Giuseppe DI Lorenzo
- Unit Oncology, Andrea Tortora Hospital, ASL Salerno, Pagani, Salerno, Italy.,Vincenzo Tiberio Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| |
Collapse
|
65
|
Møller P, Seppälä T, Dowty JG, Haupt S, Dominguez-Valentin M, Sunde L, Bernstein I, Engel C, Aretz S, Nielsen M, Capella G, Evans DG, Burn J, Holinski-Feder E, Bertario L, Bonanni B, Lindblom A, Levi Z, Macrae F, Winship I, Plazzer JP, Sijmons R, Laghi L, Valle AD, Heinimann K, Half E, Lopez-Koestner F, Alvarez-Valenzuela K, Scott RJ, Katz L, Laish I, Vainer E, Vaccaro CA, Carraro DM, Gluck N, Abu-Freha N, Stakelum A, Kennelly R, Winter D, Rossi BM, Greenblatt M, Bohorquez M, Sheth H, Tibiletti MG, Lino-Silva LS, Horisberger K, Portenkirchner C, Nascimento I, Rossi NT, da Silva LA, Thomas H, Zaránd A, Mecklin JP, Pylvänäinen K, Renkonen-Sinisalo L, Lepisto A, Peltomäki P, Therkildsen C, Lindberg LJ, Thorlacius-Ussing O, von Knebel Doeberitz M, Loeffler M, Rahner N, Steinke-Lange V, Schmiegel W, Vangala D, Perne C, Hüneburg R, de Vargas AF, Latchford A, Gerdes AM, Backman AS, Guillén-Ponce C, Snyder C, Lautrup CK, Amor D, Palmero E, Stoffel E, Duijkers F, Hall MJ, Hampel H, Williams H, Okkels H, Lubiński J, Reece J, Ngeow J, Guillem JG, Arnold J, Wadt K, Monahan K, Senter L, Rasmussen LJ, van Hest LP, Ricciardiello L, Kohonen-Corish MRJ, Ligtenberg MJL, Southey M, Aronson M, Zahary MN, Samadder NJ, Poplawski N, Hoogerbrugge N, Morrison PJ, James P, Lee G, Chen-Shtoyerman R, Ankathil R, Pai R, Ward R, Parry S, Dębniak T, John T, van Overeem Hansen T, Caldés T, Yamaguchi T, Barca-Tierno V, Garre P, Cavestro GM, Weitz J, Redler S, Büttner R, Heuveline V, Hopper JL, Win AK, Lindor N, Gallinger S, Le Marchand L, Newcomb PA, Figueiredo J, Buchanan DD, Thibodeau SN, Ten Broeke SW, Hovig E, Nakken S, Pineda M, Dueñas N, Brunet J, Green K, Lalloo F, Newton K, Crosbie EJ, Mints M, Tjandra D, Neffa F, Esperon P, Kariv R, Rosner G, Pavicic WH, Kalfayan P, Torrezan GT, Bassaneze T, Martin C, Moslein G, Ahadova A, Kloor M, Sampson JR, Jenkins MA. Colorectal cancer incidences in Lynch syndrome: a comparison of results from the prospective lynch syndrome database and the international mismatch repair consortium. Hered Cancer Clin Pract 2022; 20:36. [PMID: 36182917 PMCID: PMC9526951 DOI: 10.1186/s13053-022-00241-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 08/31/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE To compare colorectal cancer (CRC) incidences in carriers of pathogenic variants of the MMR genes in the PLSD and IMRC cohorts, of which only the former included mandatory colonoscopy surveillance for all participants. METHODS CRC incidences were calculated in an intervention group comprising a cohort of confirmed carriers of pathogenic or likely pathogenic variants in mismatch repair genes (path_MMR) followed prospectively by the Prospective Lynch Syndrome Database (PLSD). All had colonoscopy surveillance, with polypectomy when polyps were identified. Comparison was made with a retrospective cohort reported by the International Mismatch Repair Consortium (IMRC). This comprised confirmed and inferred path_MMR carriers who were first- or second-degree relatives of Lynch syndrome probands. RESULTS In the PLSD, 8,153 subjects had follow-up colonoscopy surveillance for a total of 67,604 years and 578 carriers had CRC diagnosed. Average cumulative incidences of CRC in path_MLH1 carriers at 70 years of age were 52% in males and 41% in females; for path_MSH2 50% and 39%; for path_MSH6 13% and 17% and for path_PMS2 11% and 8%. In contrast, in the IMRC cohort, corresponding cumulative incidences were 40% and 27%; 34% and 23%; 16% and 8% and 7% and 6%. Comparing just the European carriers in the two series gave similar findings. Numbers in the PLSD series did not allow comparisons of carriers from other continents separately. Cumulative incidences at 25 years were < 1% in all retrospective groups. CONCLUSIONS Prospectively observed CRC incidences (PLSD) in path_MLH1 and path_MSH2 carriers undergoing colonoscopy surveillance and polypectomy were higher than in the retrospective (IMRC) series, and were not reduced in path_MSH6 carriers. These findings were the opposite to those expected. CRC point incidence before 50 years of age was reduced in path_PMS2 carriers subjected to colonoscopy, but not significantly so.
Collapse
Affiliation(s)
- Pål Møller
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, 0379, Oslo, Norway.
| | - Toni Seppälä
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Applied Tumour Genomics Research Program, University of Helsinki, Helsinki, Finland
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - James G Dowty
- Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Saskia Haupt
- Engineering Mathematics and Computing Lab (EMCL), Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany
- Data Mining and Uncertainty Quantification (DMQ), Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | - Mev Dominguez-Valentin
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, 0379, Oslo, Norway
| | - Lone Sunde
- Department of Clinical Genetics, Aalborg University Hospital, 9000, Aalborg, Denmark
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus, Denmark
| | - Inge Bernstein
- Department of Surgical Gastroenterology, Aalborg University Hospital, Aalborg University, 9100, Aalborg, Denmark
- Institute of Clinical Medicine, Aalborg University Hospital, Aalborg University, 9100, Aalborg, Denmark
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107, Leipzig, Germany
| | - Stefan Aretz
- Institute of Human Genetics, National Center for Hereditary Tumor Syndromes, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Maartje Nielsen
- Department of Clinical Genetics, Leids Universitair Medisch Centrum, 2300RC, Leiden, The Netherlands
| | - Gabriel Capella
- Hereditary Cancer Program, Institut Català d'Oncologia-IDIBELL, L; Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Dafydd Gareth Evans
- Division of Evolution and Genomic Sciences, Manchester Centre for Genomic Medicine, University of Manchester, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - John Burn
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Elke Holinski-Feder
- Campus Innenstadt, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336, Munich, Germany
- MGZ - Center of Medical Genetics, 80335, Munich, Germany
| | - Lucio Bertario
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Zohar Levi
- Department Rabin Medical Center, Service High Risk GI Cancer Gastroenterology, Petach Tikva, Israel
| | - Finlay Macrae
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, Melbourne University, Melbourne, Australia
| | - Ingrid Winship
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, Melbourne University, Melbourne, Australia
| | | | - Rolf Sijmons
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luigi Laghi
- Department of Medicine and Surgery, Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, University of Parma, Parma, Italy
| | - Adriana Della Valle
- Hospital Fuerzas Armadas, Grupo Colaborativo Uruguayo, Investigación de Afecciones Oncológicas Hereditarias (GCU), Montevideo, Uruguay
| | - Karl Heinimann
- Medical Genetics, Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Elizabeth Half
- Gastrointestinal Cancer Prevention Unit, Gastroenterology Department, Rambam Health Care Campus, Haifa, Israel
| | | | | | - Rodney J Scott
- University of Newcastle and the Hunter Medical Research Institute, Callaghan, Australia
| | - Lior Katz
- Department of Gastroenterology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ido Laish
- The Department of Gastroenterology, High Risk and GI Cancer Prevention Clinic, Gastro-Oncology Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Elez Vainer
- Department of Gastroenterology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Carlos Alberto Vaccaro
- Hereditary Cancer Program (PROCANHE), Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Dirce Maria Carraro
- Genomic and Molecular Biology Group, A.C.Camargo Cancer Center, Sao Paulo, Brazil
| | - Nathan Gluck
- Department of Gastroenterology, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Naim Abu-Freha
- The Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aine Stakelum
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Rory Kennelly
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Des Winter
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | | | - Marc Greenblatt
- University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | | | - Harsh Sheth
- Foundation for Research in Genetics and Endocrinology, FRIGE House, Jodhpur Village Road, Satellite Ahmedabad, Ahmedabad, 380015, India
| | - Maria Grazia Tibiletti
- Ospedale di Circolo ASST Settelaghi, Centro di Ricerca Tumori Eredo-Familiari, Università dell'Insubria, Varese, Italy
| | | | - Karoline Horisberger
- Department of Abdominal and Transplantation Surgery, Universitätsspital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Carmen Portenkirchner
- Department of Abdominal and Transplantation Surgery, Universitätsspital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Ivana Nascimento
- Laboratório de Imonologia, ICS/UFBA, Núcleo de Oncologia da Bahia/Oncoclinicas, Salvador, Brazil
| | | | - Leandro Apolinário da Silva
- Hospital Universitario Oswaldo Cruz, Universidade de Pernambuco, Hospital de Câncer de Pernambuco, IPON - Instituto de Pesquisas Oncológicas do Nordeste, Salvador, Brazil
| | - Huw Thomas
- Department of Surgery and Cancer, St Mark's Hospital, Imperial College London, London, UK
| | - Attila Zaránd
- Department of Transplantation and Surgery, Semmelweis University Budapest, Budapest, Hungary
| | - Jukka-Pekka Mecklin
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- Department of Surgery, Central Finland Health Care District, Jyväskylä, Finland
| | - Kirsi Pylvänäinen
- Department of Education and Science, Central Finland Health Care District, Jyväskylä, Finland
| | - Laura Renkonen-Sinisalo
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Applied Tumour Genomics Research Program, University of Helsinki, Helsinki, Finland
| | - Anna Lepisto
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Applied Tumour Genomics Research Program, University of Helsinki, Helsinki, Finland
| | - Päivi Peltomäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Christina Therkildsen
- The Danish HNPCC Register, Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Lars Joachim Lindberg
- The Danish HNPCC Register, Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Ole Thorlacius-Ussing
- Department of Surgical Gastroenterology, Aalborg University Hospital, Aalborg University, 9100, Aalborg, Denmark
- Institute of Clinical Medicine, Aalborg University Hospital, Aalborg University, 9100, Aalborg, Denmark
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Nils Rahner
- Institute of Human Genetics, University Clinic Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Verena Steinke-Lange
- Campus Innenstadt, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336, Munich, Germany
- MGZ - Center of Medical Genetics, 80335, Munich, Germany
| | - Wolff Schmiegel
- Department of Medicine, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Deepak Vangala
- Department of Medicine, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Claudia Perne
- Institute of Human Genetics, National Center for Hereditary Tumor Syndromes, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Robert Hüneburg
- Department of Internal Medicine, University Hospital Bonn, Bonn, Germany
| | - Aída Falcón de Vargas
- Genetics Unit, Hospital Vargas de Caracas, Caracas, Venezuela
- Escuela de Medicina Jose Maria Vargas, Universidad, Central de Venezuela, UCV, Caracas, Venezuela
| | | | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, DK-2100, Copenhagen, Denmark
| | - Ann-Sofie Backman
- Department of Medicine Solna, Unit of Internal medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carmen Guillén-Ponce
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Carrie Snyder
- Hereditary Cancer Center, Department of Preventive Medicine, Creighton University, Omaha, NE, 68178, USA
| | - Charlotte K Lautrup
- Department of Clinical Genetics, Aalborg University Hospital, 9000, Aalborg, Denmark
| | - David Amor
- Murdoch Children's Research Institute and University of Melbourne Department of Paediatrics, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Edenir Palmero
- Department of Genetics, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Elena Stoffel
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Floor Duijkers
- Department of Clinical Genetics, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Heather Hampel
- Division of Human Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Heinric Williams
- Department of Urology, Geisinger Medical Center, Danville, PA, 17822, USA
| | - Henrik Okkels
- Department of Molecular Diagnostics, Aalborg University, Aalborg, Denmark
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jeanette Reece
- Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore and Cancer Genetics Service National Cancer Centre Singapore, Singapore, Singapore
| | - Jose G Guillem
- Gastrointestinal Surgery, University of North Carolina, Chapel Hill, NC, USA
| | - Julie Arnold
- New Zealand Familial Gastrointestinal Cancer Service, Auckland, New Zealand
| | - Karin Wadt
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, DK-2100, Copenhagen, Denmark
| | | | - Leigha Senter
- Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Lene J Rasmussen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Liselotte P van Hest
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Luigi Ricciardiello
- IRCCS AOU di Bologna, and Department of Medical and Surgical Sciences - University of Bologna, Bologna, Italy
| | | | - Marjolijn J L Ligtenberg
- Department of Human Genetics and Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Melissa Southey
- Monash Health Translation Precinct, Monash University, Clayton South, VIC, 3169, Australia
| | - Melyssa Aronson
- Zane Cohen Centre, Sinai Health System, Toronto, Ontario, Canada
| | - Mohd N Zahary
- Faculty of Health Sciences, University Sultan Zainal Abidin, Kuala Terengganu, Terengganu, Malaysia
| | - N Jewel Samadder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Phoenix, AZ, 85054, USA
| | - Nicola Poplawski
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patrick J Morrison
- Regional Medical Genetics Centre, Belfast HSC Trust, City Hospital Campus, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Paul James
- Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Grant Lee
- Genomics Platform Group, Centre for Cancer Research, University of Melbourne, Parkville, VIC, Australia
| | - Rakefet Chen-Shtoyerman
- The Biology Department, Ariel University, Ariel and the Oncogenetic Clinic, The Clinical Genetics Institute, Kaplan Medical Center, Rehovot, Israel
| | - Ravindran Ankathil
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Rish Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Robyn Ward
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Susan Parry
- New Zealand Familial Gastrointestinal Cancer Service, Auckland, New Zealand
| | - Tadeusz Dębniak
- Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Thomas John
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Thomas van Overeem Hansen
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, DK-2100, Copenhagen, Denmark
| | - Trinidad Caldés
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Tatsuro Yamaguchi
- Department of Clinical Genetics, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | | | - Pilar Garre
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy
| | - Jürgen Weitz
- Technische Universität Dresden, Dresden, Germany
| | - Silke Redler
- Institute of Human Genetics, University Clinic Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Reinhard Büttner
- Department of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Vincent Heuveline
- Engineering Mathematics and Computing Lab (EMCL), Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany
| | - John L Hopper
- Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Aung Ko Win
- Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Noralane Lindor
- Department of Health Science Research, Mayo Clinic Arizona, Phoenix, USA
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | | | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA
| | - Jane Figueiredo
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Stephen N Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Eivind Hovig
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, 0379, Oslo, Norway
- Department of Informatics, Centre for Bioinformatics, University of Oslo, Oslo, Norway
| | - Sigve Nakken
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, 0379, Oslo, Norway
- Centre for Cancer Cell Reprogramming (CanCell), Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marta Pineda
- Hereditary Cancer Program, Institut Català d'Oncologia-IDIBELL, L; Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Nuria Dueñas
- Hereditary Cancer Program, Institut Català d'Oncologia-IDIBELL, L; Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Joan Brunet
- Hereditary Cancer Program, Institut Català d'Oncologia-IDIBELL, L; Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Kate Green
- Division of Evolution and Genomic Sciences, Manchester Centre for Genomic Medicine, University of Manchester, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Fiona Lalloo
- Division of Evolution and Genomic Sciences, Manchester Centre for Genomic Medicine, University of Manchester, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Katie Newton
- Department of Surgery, Central Manchester University Hospitals NHS Foundation Trust and University of Manchester, Manchester, UK
| | - Emma J Crosbie
- Gynaecological Oncology Research Group, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Miriam Mints
- Division of Obstetrics and Gyneacology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Douglas Tjandra
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, Melbourne University, Melbourne, Australia
| | - Florencia Neffa
- Hospital Fuerzas Armadas, Grupo Colaborativo Uruguayo, Investigación de Afecciones Oncológicas Hereditarias (GCU), Montevideo, Uruguay
| | - Patricia Esperon
- Hospital Fuerzas Armadas, Grupo Colaborativo Uruguayo, Investigación de Afecciones Oncológicas Hereditarias (GCU), Montevideo, Uruguay
| | - Revital Kariv
- Department of Gastroenterology, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Guy Rosner
- Department of Gastroenterology, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Walter Hernán Pavicic
- Hereditary Cancer Program (PROCANHE), Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), Hospital Italiano de Buenos Aires-IUHI-CONICET, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Kalfayan
- Hereditary Cancer Program (PROCANHE), Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | | | | | - Claudia Martin
- Hospital Universitario Oswaldo Cruz, Universidade de Pernambuco, Hospital de Câncer de Pernambuco, IPON - Instituto de Pesquisas Oncológicas do Nordeste, Salvador, Brazil
| | - Gabriela Moslein
- Surgical Center for Hereditary Tumors, Ev. Bethesda Khs Duisburg, University Witten-Herdecke, Herdecke, Germany
| | - Aysel Ahadova
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Julian R Sampson
- Division of Cancer and Genetics, Institute of Medical Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Mark A Jenkins
- Centre of Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| |
Collapse
|
66
|
Zhang B, Yue J, Shi X, Cui K, Li L, Zhang C, Sun P, Zhong J, Li Z, Zhao L. Protocol of notable-HCC: a phase Ib study of neoadjuvant tislelizumab with stereotactic body radiotherapy in patients with resectable hepatocellular carcinoma. BMJ Open 2022; 12:e060955. [PMID: 36115673 PMCID: PMC9486305 DOI: 10.1136/bmjopen-2022-060955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Liver resection is the mainstay of curative-intent treatment for hepatocellular carcinoma (HCC), but the postoperative 5-year recurrence rate reaches 70%, and there are no adjuvant or neoadjuvant therapies recommended by major HCC guidelines that can reduce the risk of recurrence. In the recent decade, significant progress has been achieved in the systemic treatment of HCC, mainly from immune checkpoint inhibitors (ICIs) and targeted therapy. In other malignancies, ICIs in the neoadjuvant setting have shown better outcomes than in the adjuvant setting. On the other hand, the addition of radiation to ICIs incrementally improves the systemic response to ICIs. Neoadjuvant therapy of ICIs plus stereotactic body radiotherapy (SBRT) has shown promising results in several types of solid tumours but not HCC. METHODS AND ANALYSIS Here, we describe a phase Ib clinical trial of neoadjuvant SBRT plus PD-1 (tislelizumab) prior to hepatic resection in HCC patients. Prior to resection, eligible HCC patients will receive 8 Gy×3 fractions of SBRT together with two cycles of tislelizumab with an interval of 3 weeks. HCC resection is scheduled 4 weeks after the second dose of tislelizumab, followed by adjuvant tislelizumab for 1 year. We plan to enrol 20 participants in this trial. The primary study endpoints include the delay of surgery, tumour response and safety and tolerability of the sequential SBRT/tislelizumab. Other endpoints are the disease-free survival and overall survival rates every 3 or 6 months after the surgery. ETHICS AND DISSEMINATION This trial was approved by the Ethics Committee of Shandong Cancer Hospital and Institute (SDZLEC2022-021-01). The final results of this trial will be published in a peer-reviewed journal after completion. TRIAL REGISTRATION NUMBER NCT05185531.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinbo Yue
- Department of Abdominal Radiotherapy, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuetao Shi
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kai Cui
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lei Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chengsheng Zhang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Pengfei Sun
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhongchao Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lei Zhao
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
67
|
Dai H, Liu M, Li X, Li T, Huang W, Liao J, Li Y, Fang S. A case study of combined neoadjuvant chemotherapy and neoadjuvant immunotherapy in resectable locally advanced esophageal cancer. World J Surg Oncol 2022; 20:267. [PMID: 36008813 PMCID: PMC9414113 DOI: 10.1186/s12957-022-02732-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The prognosis of patients under existing neoadjuvant chemotherapy or neoadjuvant chemoradiotherapy requires improvement. Whereas programmed cell death 1 (PD-1) inhibitors have shown promising response in advanced esophageal cancer, they have not been used in the perioperative treatment of resectable locally advanced esophageal cancer. Whether immunotherapy can be incorporated into neoadjuvant therapy has became a challenging question for researchers. CASE PRESENTATION We present a case of a 65-year-old male who had a history of progressive dysphagia for approximately 1 month. He underwent pertinent studies including computed tomography (CT),gastroscopy,and pathological biopsy resulting in a diagnosis of medium-low differentiated squamous carcinoma of the thoracic segment of the esophagus (cT2N2M0 stage III). After 4 cycles of neoadjuvant chemotherapy combined with immunotherapy, gastroscopy showed the lesion in the esophagus was no longer present. Subsequently, the patient received thoracoscopic radical resection of esophageal cancer and achieved a pathological complete response (pCR) in postoperative pathological evaluation. During the whole treatment, no adverse effect was recorded and to date no evidence of recurrence has been recorded. CONCLUSION Our report suggest that neoadjuvant chemotherapy combined with immunotherapy not only improve the R0 resection and pCR rate in patients with resectable locally advanced esophageal cancer, but also the adverse effects are within the control range. However, the selection of therapeutic strategy, predictors of response to treatment, and interval time between neoadjuvant treatment and surgery still await more reliable evidence-based studies with large prospective samples.
Collapse
Affiliation(s)
- Huiru Dai
- The Department of Clinical Oncology, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Big data Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Minling Liu
- The Department of Clinical Oncology, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Big data Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Xueying Li
- The Department of Clinical Oncology, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Big data Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Tingwei Li
- The Department of Clinical Oncology, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Big data Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Wensheng Huang
- The Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Jiehao Liao
- The Department of Clinical Oncology, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Big data Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Yun Li
- The Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China.
| | - Shuo Fang
- The Department of Clinical Oncology, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Big data Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China.
| |
Collapse
|
68
|
Ruiz de Porras V, Pardo JC, Etxaniz O, Font A. Neoadjuvant therapy for muscle-invasive bladder cancer: Current clinical scenario, future perspectives, and unsolved questions. Crit Rev Oncol Hematol 2022; 178:103795. [PMID: 35988856 DOI: 10.1016/j.critrevonc.2022.103795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/03/2022] [Accepted: 08/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cisplatin-based neoadjuvant chemotherapy (NAC) followed by radical cystectomy is the standard treatment for patients with muscle-invasive bladder cancer (MIBC). However, the implementation of NAC is lower than desirable mainly due to its limited impact on overall survival, patients' comorbidities and the lack of predictive biomarkers to select those patients most likely to benefit from NAC. In the last decade, improved molecular MIBC characterisation, the identification of potential predictive and prognostic biomarkers as well as the incorporation of new effective therapies with a better toxicity profile, such as immunotherapy, has changed the treatment paradigm for MIBC. Therefore, the main goal for the near future is to introduce these clinical and translational advances into routine clinical practice to personalise treatment for each patient and increase the opportunity to implement bladder preservation strategies. The present review focuses on the current status of NAC in MIBC, unsolved questions and future therapeutic approaches.
Collapse
Affiliation(s)
- Vicenç Ruiz de Porras
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), 08916 Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Juan Carlos Pardo
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), 08916 Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Medical Oncology Department, Catalan Institute of Oncology, Ctra. Can Ruti - Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Olatz Etxaniz
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), 08916 Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Medical Oncology Department, Catalan Institute of Oncology, Ctra. Can Ruti - Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Albert Font
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), 08916 Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Medical Oncology Department, Catalan Institute of Oncology, Ctra. Can Ruti - Camí de les Escoles s/n, 08916 Badalona, Spain.
| |
Collapse
|
69
|
Bonadio RC, Tarantino P, Testa L, Punie K, Pernas S, Barrios C, Curigliano G, Tolaney SM, Barroso-Sousa R. Management of patients with early-stage triple-negative breast cancer following pembrolizumab-based neoadjuvant therapy: What are the evidences? Cancer Treat Rev 2022; 110:102459. [PMID: 35998514 DOI: 10.1016/j.ctrv.2022.102459] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/28/2022]
Abstract
New therapy options have changed the treatment landscape of early-stage triple-negative breast cancer (TNBC) in recent years. Most patients are candidates for neoadjuvant chemotherapy, which helps to downstage the tumor and tailor adjuvant systemic therapy based on pathologic response. Capecitabine, pembrolizumab, and olaparib have been incorporated into the armamentarium of adjuvant treatment for selected patients. The KEYNOTE-522 trial, that demonstrated the benefit of pembrolizumab, given in addition to neoadjuvant chemotherapy and adjuvantly after surgery, represented a paradigm shift for early-stage TNBC treatment. Pembrolizumab was continued in the adjuvant setting irrespective of response to neoadjuvant therapy, and other adjuvant therapies were not administered in the trial. Many questions were then raised on the selection of adjuvant therapy regimens for patients with residual disease (RD). Prior to the routine use of immune-checkpoint inhibitors (ICI), the value of adjuvant capecitabine for patients with RD after neoadjuvant polychemotherapy was demonstrated. Given the poor prognosis of some patients with RD after neoadjuvant chemo-immunotherapy, while the survival advantage of adding capecitabine during the adjuvant phase of pembrolizumab is unknown, it does appear safe and can be considered. Regarding patients harboring germline BRCA mutations with RD after neoadjuvant ICI-containing regimens, the combination of olaparib with pembrolizumab can be an option based on existing safety data.
Collapse
Affiliation(s)
- Renata Colombo Bonadio
- Instituto D'Or de Pesquisa e Ensino (IDOR), São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; European Institute of Oncology, IRCCS
| | - Laura Testa
- Instituto D'Or de Pesquisa e Ensino (IDOR), São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Sonia Pernas
- Catalan Institute of Oncology; IDIBELL, L'Hospitalet de Llobregat (Barcelona), Spain
| | - Carlos Barrios
- Latin American Cooperative Oncology Group (LACOG), Grupo Oncoclínicas, Porto Alegre, Brazil
| | | | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Romualdo Barroso-Sousa
- Oncology Center, Hospital Sirio-Libanes, Brasília, Brazil; DASA Oncology, Brasília, Brazil.
| |
Collapse
|
70
|
Rohaan MW, Stahlie EHA, Franke V, Zijlker LP, Wilgenhof S, van der Noort V, van Akkooi ACJ, Haanen JBAG. Neoadjuvant nivolumab + T-VEC combination therapy for resectable early stage or metastatic (IIIB-IVM1a) melanoma with injectable disease: study protocol of the NIVEC trial. BMC Cancer 2022; 22:851. [PMID: 35927710 PMCID: PMC9351098 DOI: 10.1186/s12885-022-09896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/13/2022] [Indexed: 11/27/2022] Open
Abstract
Background Trials investigating neoadjuvant treatment with immune checkpoint inhibitors (ICI) in patients with melanoma have shown high clinical and pathologic response rates. Treatment with talimogene laherparepvec (T-VEC), a modified herpes simplex virus type-1 (HSV-1), is approved for patients with unresectable stage IIIB-IVM1a melanoma and has the potential to make tumors more susceptible for ICI. Combination ICI and intralesional T-VEC has already been investigated in patients with unresectable stage IIIB-IV disease, however, no data is available yet on the potential benefit of this combination therapy in neoadjuvant setting. Methods This single center, single arm, phase II study aims to show an improved major pathologic complete response (pCR) rate, either pCR or near-pCR, up to 45% in 24 patients with resectable stage IIIB-IVM1a melanoma upon neoadjuvant combination treatment with intralesional T-VEC and systemic nivolumab (anti-PD-1 antibody). Patients will receive four courses of T-VEC up to 4 mL (first dose as seroconversion dose) and three doses of nivolumab (240 mg flatdose) every 2 weeks, followed by surgical resection in week nine. The primary endpoint of this trial is pathologic response rate. Secondary endpoints are safety, the rate of delay of surgery and event-free survival. Additionally, prognostic and predictive biomarker research and health-related quality of life evaluation will be performed. Discussion Intralesional T-VEC has the capacity to heighten the immune response and to elicit an abscopal effect in melanoma in combination with ICI. However, the potential clinical benefit of T-VEC plus ICI in the neoadjuvant setting remains unknown. This is the first trial investigating the efficacy and safety of neoadjuvant treatment of T-VEC and nivolumab followed by surgical resection in patients with stage IIIB-IVM1a melanoma, with the potential of high pathologic response rates and acceptable toxicity. Trial registration This trial was registered in the European Union Drug Regulating Authorities Clinical Trials Database (EudraCT- number: 2019–001911-22) and the Central Committee on Research Involving Human Subjects (NL71866.000.19) on 4th June 2020. Secondary identifying number: NCT04330430.
Collapse
Affiliation(s)
- Maartje W Rohaan
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Emma H A Stahlie
- Department of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Viola Franke
- Department of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Lisanne P Zijlker
- Department of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Vincent van der Noort
- Department of Biometrics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands.
| |
Collapse
|
71
|
Xiao J, Liu T, Liu Z, Xiao C, Du J, Zuo S, Li H, Gu H. A Differentiation-Related Gene Prognostic Index Contributes to Prognosis and Immunotherapy Evaluation in Patients with Hepatocellular Carcinoma. Cells 2022; 11:cells11152302. [PMID: 35892599 PMCID: PMC9367442 DOI: 10.3390/cells11152302] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common gastrointestinal tumor with a poor prognosis, which is associated with poor differentiation of tumor cells. However, the potential value of cell differentiation-related molecules in predicting the benefit and prognosis of immune checkpoint inhibitors (ICI) therapy remains unknown. Herein, to investigate the differentiation trajectory of HCC cells and their clinical significance, a differentiation-related gene prognostic index (DRGPI) based on HCC differentiation-related genes (HDRGs) was constructed to elucidate the immune characteristics and therapeutic benefits of ICI in the HCC subgroup defined by DRGPI. Single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data from four HCC samples were integrated for bioinformatics analysis. Then, PON1, ADH4, SQSTM1, HSP90AA1, and STMN1 were screened out to construct a DRGPI. More intriguingly, RT-qPCR validation of the expression of these genes yielded consistent results with the TCGA database. Next, the risk scoring (RS) constructed based on DRGPI suggested that the overall survival (OS) of the DRGPI-high patients was significantly worse than that of the DRGPI-low patients. A nomogram was constructed based on DRGPI-RS and clinical characteristics, which showed strong predictive performance and high accuracy. The comprehensive results indicated that a low DRGPI score was associated with low TP53 mutation rates, high CD8 T cell infiltration, and more benefit from ICI therapy. Homoplastically, the high DRGPI score reflected the opposite results. Taken together, our study highlights the significance of HCC cell differentiation in predicting prognosis, indicating immune characteristics, and understanding the therapeutic benefits of ICI, and suggests that DRGPI is a valuable prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Jingjing Xiao
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
- Department of Hepatobiliary Surgery, Guizhou Provincial People’s Hospital, Guiyang 550002, China;
- Department of Pediatric Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Tao Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
- Department of Hepatobiliary Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, China
| | - Zhenhua Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People’s Hospital, Guiyang 550002, China;
| | - Chuan Xiao
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
| | - Jun Du
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
- Department of Pediatric Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Shi Zuo
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Haiyang Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Huajian Gu
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, China; (J.X.); (T.L.); (C.X.); (J.D.); (S.Z.); (H.L.)
- Department of Pediatric Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
- Correspondence: ; Tel.: +86-851-8677-2723
| |
Collapse
|
72
|
Roviello G, Catalano M, Santi R, Santoni M, Galli IC, Amorosi A, Polom W, De Giorgi U, Nesi G. Neoadjuvant Treatment in Muscle-Invasive Bladder Cancer: From the Beginning to the Latest Developments. Front Oncol 2022; 12:912699. [PMID: 35936721 PMCID: PMC9353067 DOI: 10.3389/fonc.2022.912699] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Urothelial carcinoma of the bladder is one of the most prevalent cancers worldwide, diagnosed as muscle invasive in 25% of cases. Although several studies have demonstrated an overall 5% absolute survival benefit at 5 years with cisplatin-based combination neoadjuvant treatment, administration of chemotherapy prior to radical cystectomy (RC) in muscle-invasive bladder cancer (MIBC) patients is still a matter of debate. This may be due to the perceived modest survival benefit, cisplatin-based chemotherapy ineligibility, or fear of delaying potentially curative surgery in non-responders. However, immunotherapy and novel targeted therapies have shown to prolong survival in advanced disease and are under investigation in the neoadjuvant and adjuvant settings to reduce systemic relapse and improve cure rates. Genomic characterization of MIBC could help select the most effective chemotherapeutic regimen for the individual patient. Large cohort studies on neoadjuvant treatments with immune checkpoint inhibitors (ICIs) and molecular therapies, alone or combined with chemotherapy, are ongoing. In this review, we trace the development of neoadjuvant therapy in MIBC and explore recent advances that may soon change clinical practice.
Collapse
Affiliation(s)
| | - Martina Catalano
- School of Human Health Sciences, University of Florence, Florence, Italy
| | - Raffaella Santi
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Ilaria Camilla Galli
- Histopathology and Molecular Diagnostics, Careggi Teaching Hospital, Florence, Italy
| | - Andrea Amorosi
- Department of Health Sciences, University of Catanzaro, Catanzaro, Italy
| | - Wojciech Polom
- Department of Urology, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, Meldola, Italy
| | - Gabriella Nesi
- Department of Health Sciences, University of Florence, Florence, Italy
- *Correspondence: Gabriella Nesi,
| |
Collapse
|
73
|
Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell 2022; 185:2918-2935.e29. [PMID: 35803260 PMCID: PMC9508682 DOI: 10.1016/j.cell.2022.06.018] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/26/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022]
Abstract
Neoadjuvant immune checkpoint blockade has shown promising clinical activity. Here, we characterized early kinetics in tumor-infiltrating and circulating immune cells in oral cancer patients treated with neoadjuvant anti-PD-1 or anti-PD-1/CTLA-4 in a clinical trial (NCT02919683). Tumor-infiltrating CD8 T cells that clonally expanded during immunotherapy expressed elevated tissue-resident memory and cytotoxicity programs, which were already active prior to therapy, supporting the capacity for rapid response. Systematic target discovery revealed that treatment-expanded tumor T cell clones in responding patients recognized several self-antigens, including the cancer-specific antigen MAGEA1. Treatment also induced a systemic immune response characterized by expansion of activated T cells enriched for tumor-infiltrating T cell clonotypes, including both pre-existing and emergent clonotypes undetectable prior to therapy. The frequency of activated blood CD8 T cells, notably pre-treatment PD-1-positive KLRG1-negative T cells, was strongly associated with intra-tumoral pathological response. These results demonstrate how neoadjuvant checkpoint blockade induces local and systemic tumor immunity.
Collapse
|
74
|
Ho G, Schwartz RJ, Regio Pereira A, Dimitrou F, Paver E, McKenzie C, Saw RPM, Scolyer RA, Long GV, Guitera P. Reflectance confocal microscopy - a non-invasive tool for monitoring systemic treatment response in stage III unresectable primary scalp melanoma. J Eur Acad Dermatol Venereol 2022; 36:e583-e585. [PMID: 35285090 DOI: 10.1111/jdv.18076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/04/2022] [Indexed: 11/27/2022]
Affiliation(s)
- G Ho
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - R J Schwartz
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Dermatology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - A Regio Pereira
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Melanoma Diagnostic Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Federal University of Sao Paulo, Sao Paulo, Brazil
| | - F Dimitrou
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - E Paver
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - C McKenzie
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - R P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - R A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - G V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital and Mater Hospitals, Sydney, New South Wales, Australia
| | - P Guitera
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Melanoma Diagnostic Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
75
|
Kovács SA, Győrffy B. Transcriptomic datasets of cancer patients treated with immune-checkpoint inhibitors: a systematic review. J Transl Med 2022; 20:249. [PMID: 35641998 PMCID: PMC9153191 DOI: 10.1186/s12967-022-03409-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/24/2022] [Indexed: 12/12/2022] Open
Abstract
The availability of immune-checkpoint inhibitors (ICI) in the last decade has resulted in a paradigm shift in certain areas of oncology. Patients can be treated either by a monotherapy of anti-CTLA-4 (tremelimumab or ipilimumab), anti-PD-1 (nivolumab or pembrolizumab), or anti-PD-L1 (avelumab or atezolizumab or durvalumab) or as combination therapy of anti-CTLA-4 and anti-PD-1. To maximize the clinical treatment benefit of cancer immunotherapy, the prediction of the actual immune response by the identification and application of clinically useful biomarkers will be required. Whole transcriptomic datasets of patients with ICI treatment could provide the basis for large-scale discovery and ranking of such potential biomarker candidates. In this review, we summarize currently available transcriptomic data from different biological sources (whole blood, fresh-frozen tissue, FFPE) obtained by different methods (microarray, RNA-Seq, RT-qPCR). We directly include only results from clinical trials and other investigations where an ICI treatment was administered. The available datasets are grouped based on the administered treatment and we also summarize the most important results in the individual cohorts. We discuss the limitations and shortcomings of the available datasets. Finally, a subset of animal studies is reviewed to provide an overview of potential in vivo ICI investigations. Our review can provide a swift reference for researchers aiming to find the most suitable study for their investigation, thus saving a significant amount of time.
Collapse
Affiliation(s)
- Szonja Anna Kovács
- grid.11804.3c0000 0001 0942 9821Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094 Budapest, Hungary ,grid.429187.10000 0004 0635 9129Research Centre for Natural Sciences, Oncology Biomarker Research Group, Institute of Enzymology, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, 1117 Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary. .,Research Centre for Natural Sciences, Oncology Biomarker Research Group, Institute of Enzymology, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, 1117, Budapest, Hungary.
| |
Collapse
|
76
|
Aguado C, Chara L, Antoñanzas M, Matilla Gonzalez JM, Jiménez U, Hernanz R, Mielgo-Rubio X, Trujillo-Reyes JC, Couñago F. Neoadjuvant treatment in non-small cell lung cancer: New perspectives with the incorporation of immunotherapy. World J Clin Oncol 2022; 13:314-322. [PMID: 35662985 PMCID: PMC9153074 DOI: 10.5306/wjco.v13.i5.314] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/24/2021] [Accepted: 05/07/2022] [Indexed: 02/06/2023] Open
Abstract
The aim of neoadjuvant treatment in non-small cell lung cancer (NSCLC) is to eliminate micrometastatic disease to facilitate surgical resection. Neoadjuvant chemotherapy (ChT) in localised NSCLC has numerous advantages over other therapeutic modalities and is considered standard treatment in resectable disease. Treatment with immune checkpoint inhibitors (ICI) improves long-term survival in advanced disease and has a better toxicity profile than conventional therapies. These immunotherapy agents (anti-PD1/PD-L1), administered with or without ChT, are currently being evaluated in the preoperative setting, with initial results showing better pathological response rates and more long-term benefits. Importantly, these drugs do not appear to increase the rate of severe adverse effects and/or postoperative complications. However, several questions still need to be resolved, including the identification of predictive biomarkers; comparative studies of immunotherapy alone vs combined treatment with ChT and/or radiotherapy; the optimal duration of treatment; the timing of surgery; the need for adjuvant treatment; appropriate radiologic evaluation and mediastinal staging; and the correlation between pathological response and survival outcomes. Here we review the current evidence for immunotherapy from a multidisciplinary perspective and discuss current and future controversies.
Collapse
Affiliation(s)
- Carlos Aguado
- Department of Medical Oncology, Hospital Clínico Universitario San Carlos, Madrid 28040, Spain
| | - Luis Chara
- Department of Medical Oncology, Hospital Universitario de Guadalajara, Guadalajara 19002, Spain
| | - Mónica Antoñanzas
- Department of Medical Oncology, Hospital Clínico Universitario San Carlos, Madrid 28040, Spain
| | | | - Unai Jiménez
- Department of Thoracic Surgery, Hospital Universitario Cruces, Barakaldo, Bizkaia 48903, Basque Country, Spain
| | - Raul Hernanz
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid 28034, Spain
| | - Xabier Mielgo-Rubio
- Department of Medical Oncology, Hospital Universitario Fundación Alcorcón, Alcorcón 28922, Madrid, Spain
| | - Juan Carlos Trujillo-Reyes
- Department of Thoracic Surgery, Hospital de la Santa Creu I Sant Pau, Barcelona 08029, Catalonia, Spain
- Department of Surgery, Universitat Autonoma de Barcelona, Barcelona 08029, Catalonia, Spain
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario Quirónsalud Madrid, Pozuelo de Alarcón 28223, Madrid, Spain
- Department of Radiation Oncology, Hospital La Luz, Madrid 28003, Spain
- Medicine Department, School of Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón 28670, Madrid, Spain
| |
Collapse
|
77
|
Petrelli F, Ghidini A, Simioni A, Campana LG. Impact of electrochemotherapy in metastatic cutaneous melanoma: a contemporary systematic review and meta-analysis. Acta Oncol 2022; 61:533-544. [PMID: 34889156 DOI: 10.1080/0284186x.2021.2006776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Electrochemotherapy (ECT) harnesses electric pulses to enhance cytotoxic drug delivery into tumors and has entered the armamentarium to treat superficially metastatic melanoma. We performed a systematic review and meta-analysis to assess treatment patterns and patient outcomes. METHODS PubMed, Medline, Embase, and the Cochrane Library databases were queried for publication from inception to September 2020. Primary outcome measures were overall and complete response rate (ORR and CRR); secondary outcomes included local control rate (LCR) and overall survival (OS). RESULTS Twenty-seven studies met the selection criteria for a total of 1161 individuals (mean age 71 years) and 5308 tumors (weighted mean size 14 mm). The majority of patients (n = 1124) underwent bleomycin-ECT. Aggregate ORR was 77.6% (95% confidence interval [CI] 71.0 - 83.2%) and CRR 48% (95% CI 42 - 54%), with no significant difference between the route of bleomycin administration (ORR, 69.2 vs. 81.9% following intravenous or intratumoral bleomycin, p = .37) and tumor size (p = .69). When reported (n = 8 studies), 1- and 2-year LCR ranged from 54 to 89% and 72 to 74%, respectively, and 1-year OS (n = 3 studies) from 67 to 89%. CONCLUSIONS ECT with either intratumoral or intravenous bleomycin confers a high therapeutic response in cutaneous metastatic melanoma. Moderate evidence supports its low toxicity and durability of local control.HighlightsElectrochemotherapy (ECT) is associated with a 77% overall response rate (ORR).Intravenous and intratumoral bleomycin are equally effective.There are no relevant toxicity concerns.One-year local tumor control rate ranges from 54 to 89%.Current literature has significant variation in reporting.
Collapse
Affiliation(s)
| | | | - Andrea Simioni
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
78
|
André T, Cohen R, Salem ME. Immune Checkpoint Blockade Therapy in Patients With Colorectal Cancer Harboring Microsatellite Instability/Mismatch Repair Deficiency in 2022. Am Soc Clin Oncol Educ Book 2022; 42:1-9. [PMID: 35471834 DOI: 10.1200/edbk_349557] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to be effective among patients with metastatic colorectal cancer (mCRC) harboring high microsatellite instability (MSI-H) and/or mismatch repair deficiency (dMMR), with U.S. Food and Drug Administration approvals for all lines of therapy. In Europe, only pembrolizumab in the first line and the combination of nivolumab and ipilimumab beyond the first line are approved. Many questions remain about the clinical management of MSI-H/dMMR CRC. Biomarkers predictive of immune checkpoint inhibitor resistance among MSI-H/dMMR tumors are needed (1) to select the best treatment for patients with CRC (anti-PD-[L]1 monotherapy alone or combined with anti-CTLA-4 or chemotherapy) and (2) to develop new treatment strategies for patients whose disease progressed after immune checkpoint inhibitor monotherapy. The development of immune checkpoint inhibitors in the adjuvant and neoadjuvant settings is also of great interest for patients harboring MSI-H/dMMR, especially as a substantial proportion have Lynch syndrome or are at high risk of developing cancers in their lifetime and sporadic MSI-H/dMMR cancers occur most frequently in elderly and frail patients. Thus, CRC is not one, but two different diseases: (1) MSI-H/dMMR CRC (seen in 5% of mCRC and 15% of non-mCRC), which is genetically unstable with a high mutational load and many neoantigens, and for which immune checkpoint inhibitors radically changed clinical management, and (2) microsatellite stable CRC with chromosomal instability, for which immune checkpoint inhibitors are not proven efficient.
Collapse
Affiliation(s)
- Thierry André
- Sorbonne University, Saint-Antoine Hospital, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S938, Paris, France
| | - Romain Cohen
- Sorbonne University, Saint-Antoine Hospital, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S938, Paris, France
| | | |
Collapse
|
79
|
Kaptein P, Jacoberger-Foissac C, Dimitriadis P, Voabil P, de Bruijn M, Brokamp S, Reijers I, Versluis J, Nallan G, Triscott H, McDonald E, Tay J, Long GV, Blank CU, Thommen DS, Teng MWL. Addition of interleukin-2 overcomes resistance to neoadjuvant CTLA4 and PD1 blockade in ex vivo patient tumors. Sci Transl Med 2022; 14:eabj9779. [PMID: 35476594 DOI: 10.1126/scitranslmed.abj9779] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neoadjuvant immunotherapy with anti-cytotoxic T lymphocyte-associated protein 4 (CTLA4) + anti-programmed cell death protein 1 (PD1) monoclonal antibodies has demonstrated remarkable pathological responses and relapse-free survival in ~80% of patients with clinically detectable stage III melanoma. However, about 20% of the treated patients do not respond. In pretreatment biopsies of patients with melanoma, we found that resistance to neoadjuvant CTLA4 + PD1 blockade was associated with a low CD4/interleukin-2 (IL-2) gene signature. Ex vivo, addition of IL-2 to CTLA4 + PD1 blockade induced T cell activation and deep immunological responses in anti-CTLA4 + anti-PD1-resistant human tumor specimens. In the 4T1.2 breast cancer mouse model of neoadjuvant immunotherapy, triple combination of anti-CTLA4 + anti-PD1 + IL-2 cured almost twice as many mice as compared with dual checkpoint inhibitor therapy. This improved efficacy was due to the expansion of tumor-specific CD8+ T cells and improved proinflammatory cytokine polyfunctionality of both CD4+ and CD8+ T effector cells and regulatory T cells. Depletion studies suggested that CD4+ T cells were critical for priming of CD8+ T cell immunity against 4T1.2 and helped in the expansion of tumor-specific CD8+ T cells early after neoadjuvant triple immunotherapy. Our results suggest that the addition of IL-2 can overcome resistance to neoadjuvant anti-CTLA4 + anti-PD1, providing the rationale for testing this combination as a neoadjuvant therapy in patients with early-stage cancer.
Collapse
Affiliation(s)
- Paulien Kaptein
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | | | - Petros Dimitriadis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Paula Voabil
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Marjolein de Bruijn
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Simone Brokamp
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Irene Reijers
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Judith Versluis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Gahyathiri Nallan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Hannah Triscott
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia.,School of Medicine, University of Queensland, Herston, Queensland 4006, Australia
| | - Elizabeth McDonald
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Joshua Tay
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney 2006, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia.,Royal North Shore and Mater Hospitals, Sydney 2065, Australia
| | - Christian U Blank
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Daniela S Thommen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Michele W L Teng
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia.,School of Medicine, University of Queensland, Herston, Queensland 4006, Australia
| |
Collapse
|
80
|
Rocha P, Zhang J, Laza-Briviesca R, Cruz-Bermúdez A, Bota-Rabassedas N, Sanchez-Espiridion B, Yoshimura K, Behrens C, Lu W, Tang X, Pataer A, Parra ER, Haymaker C, Fujimoto J, Swisher SG, Heymach JV, Gibbons DL, Lee JJ, Sepesi B, Cascone T, Solis LM, Provencio M, Wistuba II, Kadara H. Distinct immune gene programs associated with host tumor immunity, neoadjuvant chemotherapy and chemoimmunotherapy in resectable NSCLC. Clin Cancer Res 2022; 28:2461-2473. [PMID: 35394499 DOI: 10.1158/1078-0432.ccr-21-3207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 02/12/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Our understanding of the immunopathology of resectable NSCLC is still limited. Here, we explore immune programs that inform of tumor immunity and response to neoadjuvant chemotherapy and chemoimmunotherapy in localized NSCLC. EXPERIMENTAL DESIGN Targeted immune gene sequencing using the HTG Precision Immuno-Oncology panel was performed in localized NSCLCs from three cohorts based on treatment: naïve (n=190), neoadjuvant chemotherapy (n=38) and neoadjuvant chemoimmunotherapy (n=21). Tumor immune microenvironment (TIME) phenotypes based on the location of CD8+ T cells (inflamed, cold, excluded), tumoral PD-L1 expression (<1% and {greater than or equal to}1%), and tumor infiltrating lymphocytes (TILs). Immune programs and signatures were statistically analyzed based on tumoral PD-L1 expression, immune phenotypes, pathological response and were cross-compared across the three cohorts. RESULTS PD-L1 positive tumors exhibited increased signature scores for various lymphoid and myeloid cell subsets (p<0.05). TIME phenotypes exhibited disparate frequencies by stage, PD-L1 expression, and mutational burden. Inflamed and PD-L1+/TILs+ NSCLCs displayed overall significantly heightened levels of immune signatures, with the excluded group representing an intermediate state. A cytotoxic T cell signature was associated with favorable survival in neoadjuvant chemotherapy-treated NSCLCs (p<0.05). Pathological response to chemoimmunotherapy was positively associated with higher expression of genes involved in immune activation, chemotaxis, as well as T and NK cells (p<0.05 for all). Among the three cohorts, chemoimmunotherapy-treated NSCLCs exhibited highest scores for various immune cell subsets including T effector and B cells (p<0.05). CONCLUSIONS Our findings highlight immune gene programs that may underlie host tumor immunity and response to neoadjuvant chemotherapy and chemoimmunotherapy in resectable NSCLC.
Collapse
Affiliation(s)
- Pedro Rocha
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Jiexin Zhang
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | - Alberto Cruz-Bermúdez
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain., Madrid, Spain
| | | | | | - Katsuhiro Yoshimura
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Carmen Behrens
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wei Lu
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ximing Tang
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Apar Pataer
- The University of Texas MD Anderson Cancer Center, houston, Texas, United States
| | - Edwin R Parra
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cara Haymaker
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Junya Fujimoto
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen G Swisher
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John V Heymach
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Don L Gibbons
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - J Jack Lee
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Boris Sepesi
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tina Cascone
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M Solis
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mariano Provencio
- Medical Oncology Department, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain., Majadahonda, Madrid, Spain
| | - Ignacio I Wistuba
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Humam Kadara
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
81
|
Ju S, Zhou C, Hu J, Wang Y, Wang C, Liu J, Yang C, Huang S, Li T, Chen Y, Bai Y, Yao W, Xiong B. Late combination of transarterial chemoembolization with apatinib and camrelizumab for unresectable hepatocellular carcinoma is superior to early combination. BMC Cancer 2022; 22:335. [PMID: 35346114 PMCID: PMC8961945 DOI: 10.1186/s12885-022-09451-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/22/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Objective
The purpose of this study was to explore the efficacy and safety of transarterial chemoembolization (TACE) combined with apatinib and camrelizumab (TACE + AC) for unresectable hepatocellular carcinoma (HCC), and the impact of the timing of the combination on it.
Methods
In this single-arm retrospective study, consecutive data of patients with unresectable HCC treated to our hospital from March 2017 to September 2021 were collected. These patients were treated with TACE and started on camrelizumab and apatinib within one week of TACE. Camrelizumab 200 mg intravenously once every three weeks and apatinib 250 mg orally once daily. Repeat TACE treatment was available on an on-demand basis. The primary endpoints were overall survival (OS) and progression-free survival (PFS). Secondary endpoints included objective response rate (ORR), disease control rate (DCR), and safety. The univariate and multivariate Cox regression analyses were used to assess the effect of early and late combination on OS and PFS.
Results
A total of 80 patients were enrolled in this study. The median OS was 22.1 months (95% confidence interval [CI]: 13.8–30.5 months) and the median PFS was 15.7 months (95% CI: 14.7–16.6 months). The ORR was 58.8% (95% CI: 47.2–69.6) and DCR reached 81.2% (95% CI: 71.0–89.1). Multivariable Cox proportional hazard regression analyses showed that TACE late combined with apatinib and camrelizumab provided better OS than early combination (HR = 0.175, 95% CI:0.060–0.509, P = 0.001), as did PFS (HR = 0.422, 95% CI:0.184–0.967, P = 0.041). All treatment-related adverse events were tolerable, and no serious adverse events were observed.
Conclusion
TACE combined with apatinib plus camrelizumab for patients with unresectable HCC has promising antitumor activity and a manageable safety profile. For unresectable HCC with large tumor burden, late combination provides better OS and PFS compared to early combination.
Collapse
|
82
|
Siebenhüner AR, Langheinrich M, Friemel J, Schaefer N, Eshmuminov D, Lehmann K. Orchestrating Treatment Modalities in Metastatic Pancreatic Neuroendocrine Tumors-Need for a Conductor. Cancers (Basel) 2022; 14:1478. [PMID: 35326628 PMCID: PMC8946777 DOI: 10.3390/cancers14061478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are a vast growing disease. Over 50% of these tumors are recognized at advanced stages with lymph node, liver, or distant metastasis. An ongoing controversy is the role of surgery in the metastatic setting as dedicated systemic treatments have emerged recently and shown benefits in randomized trials. Today, liver surgery is an option for advanced pNETs if the tumor has a favorable prognosis, reflected by a low to moderate proliferation index (G1 and G2). Surgery in this well-selected population may prolong progression-free and overall survival. Optimal selection of a treatment plan for an individual patient should be considered in a multidisciplinary tumor board. However, while current guidelines offer a variety of modalities, there is so far only a limited focus on the right timing. Available data is based on small case series or retrospective analyses. The focus of this review is to highlight the right time-point for surgery in the setting of the multimodal treatment of an advanced pancreatic neuroendocrine tumor.
Collapse
Affiliation(s)
- Alexander R. Siebenhüner
- Clinic for Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
- ENETS Center of Excellence Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland;
| | - Melanie Langheinrich
- Department of Visceral Surgery, University Hospital Greifswald, Ferdinand-Sauerbruch-Strasse, D-17475 Greifswald, Germany;
| | - Juliane Friemel
- Institute for Pathologie, University Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland;
| | - Niklaus Schaefer
- Department of Nuclear Medicine, University Hospital Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland;
| | - Dilmurodjon Eshmuminov
- Department of Surgery and Transplantation, University Hospital of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland;
| | - Kuno Lehmann
- ENETS Center of Excellence Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland;
- Department of Surgery and Transplantation, University Hospital of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland;
| |
Collapse
|
83
|
Witt RG, Erstad DJ, Wargo JA. Neoadjuvant therapy for melanoma: rationale for neoadjuvant therapy and pivotal clinical trials. Ther Adv Med Oncol 2022; 14:17588359221083052. [PMID: 35251322 PMCID: PMC8894940 DOI: 10.1177/17588359221083052] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/03/2022] [Indexed: 12/26/2022] Open
Abstract
The treatment of malignant melanoma has drastically changed over the past decade with the advent of immune checkpoint blockade, targeted therapy with BRAF/MEK inhibition, and other novel therapies such as oncolytic virus intralesional therapy. Despite improvements in patient response rates and survival with these new treatments, there exists a large portion of patients with surgically resectable disease that are high risk for relapse. Patients with high-risk resectable melanoma account for up to 20% of newly diagnosed cases. For this high-risk group of patients, neoadjuvant therapy has many purposed advantages over adjuvant therapy, including a more robust immune response due to abundant tumor antigens at treatment initiation, the ability to assess pathologic response to therapy, tumor downstaging leading to increased disease resectability, and a potential decreased need for extensive lymphadenectomies. These findings have been backed by preclinical models and multiple neoadjuvant trials are underway. In this review, we will discuss the trials that have set the foundation for the current treatment standards and discuss the role and rationale for neoadjuvant therapy for high-risk malignant melanomas.
Collapse
Affiliation(s)
- Russell G. Witt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Derek J. Erstad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1484, Houston, TX 77030-4009, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
84
|
Wu D, Huang H, Zhang M, Li Z, Wang S, Yu Y, Fang Y, Jiang N, Miao H, Ma P, Tang Y, Li N. The global landscape of neoadjuvant and adjuvant anti-PD-1/PD-L1 clinical trials. J Hematol Oncol 2022; 15:16. [PMID: 35135567 PMCID: PMC8822713 DOI: 10.1186/s13045-022-01227-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
The neoadjuvant and adjuvant anti-PD-1/PD-L1 treatment has been increasingly noticed. To summarize the global landscape of these clinical trials will provide essential data for all the stakeholders of drug development. Based on the Trialtrove database, a total of 668 clinical trials initiated by the end of 2020 were retrospectively analyzed. We found that a rising capability of global neoadjuvant and adjuvant anti-PD-1/PD-L1 clinical development has been achieved. High prevalent cancer types were extensively studied though the priorities in China and the United States were different. However, a lack of phase III trials and industry-sponsored trials was addressed. The confirmatory neoadjuvant trials were particularly insufficient, and the combination strategy mainly focused on chemotherapy. Thus, more public funding and accelerated regulatory strategies are needed in this field. Efforts should be made to confirm the benefit of neoadjuvant treatment and explore novel combination strategies.
Collapse
Affiliation(s)
- Dawei Wu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huiyao Huang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ziwei Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shuhang Wang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Yu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Fang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Jiang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huilei Miao
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peiwen Ma
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Tang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
85
|
Du F, Peng L, Wang Q, Dong K, Pei W, Zhuo H, Xu T, Jing C, Li L, Zhang J. CCDC12 promotes tumor development and invasion through the Snail pathway in colon adenocarcinoma. Cell Death Dis 2022; 13:187. [PMID: 35217636 PMCID: PMC8881494 DOI: 10.1038/s41419-022-04617-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/10/2022] [Accepted: 02/09/2022] [Indexed: 12/02/2022]
Abstract
Integrative expression Quantitative Trait Loci (eQTL) analysis found that rs8180040 was significantly associated with Coiled-coil domain containing 12 (CCDC12) in colon adenocarcinoma (COAD) patients. Immunohistochemical staining and western blotting confirmed CCDC12 was highly expressed in COAD tissues, which was consistent with RNA-Seq data from the TCGA database. Knockdown of CCDC12 could significantly reduce proliferation, migration, invasion, and tumorigenicity of colon cancer cells, while exogenous overexpression of CCDC12 had the opposite effect. Four plex Isobaric Tags for Relative and Absolute Quantitation assays were performed to determine its function and potential regulatory mechanism and demonstrated that overexpression of CCDC12 would change proteins on the adherens junction pathway. Overexpressed Snail and knocked down CCDC12 subsequently in SW480 cells, and we found that overexpression of Snail did not significantly change CCDC12 levels in SW480 cells, while knockdown of CCDC12 reduced that of Snail. CCDC12 plays a significant role in tumorigenesis, development, and invasion of COAD and may affect the epithelial to mesenchymal transformation process of colon cancer cells by regulating the Snail pathway.
Collapse
|
86
|
Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov 2021; 21:509-528. [PMID: 34937915 DOI: 10.1038/s41573-021-00345-8] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/11/2022]
Abstract
Cancer immunity, and the potential for cancer immunotherapy, have been topics of scientific discussion and experimentation for over a hundred years. Several successful cancer immunotherapies - such as IL-2 and interferon-α (IFNα) - have appeared over the past 30 years. However, it is only in the past decade that immunotherapy has made a broad impact on patient survival in multiple high-incidence cancer indications. The emergence of immunotherapy as a new pillar of cancer treatment (adding to surgery, radiation, chemotherapy and targeted therapies) is due to the success of immune checkpoint blockade (ICB) drugs, the first of which - ipilimumab - was approved in 2011. ICB drugs block receptors and ligands involved in pathways that attenuate T cell activation - such as cytotoxic T lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1) and its ligand, PDL1 - and prevent, or reverse, acquired peripheral tolerance to tumour antigens. In this Review we mark the tenth anniversary of the approval of ipilimumab and discuss the foundational scientific history of ICB, together with the history of the discovery, development and elucidation of the mechanism of action of the first generation of drugs targeting the CTLA4 and PD1 pathways.
Collapse
|
87
|
Karin N. Chemokines in the Landscape of Cancer Immunotherapy: How They and Their Receptors Can Be Used to Turn Cold Tumors into Hot Ones? Cancers (Basel) 2021; 13:6317. [PMID: 34944943 PMCID: PMC8699256 DOI: 10.3390/cancers13246317] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last decade, monoclonal antibodies to immune checkpoint inhibitors (ICI), also known as immune checkpoint blockers (ICB), have been the most successful approach for cancer therapy. Starting with mAb to cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors in metastatic melanoma and continuing with blockers of the interactions between program cell death 1 (PD-1) and its ligand program cell death ligand 1 (PDL-1) or program cell death ligand 2 (PDL-2), that have been approved for about 20 different indications. Yet for many cancers, ICI shows limited success. Several lines of evidence imply that the limited success in cancer immunotherapy is associated with attempts to treat patients with "cold tumors" that either lack effector T cells, or in which these cells are markedly suppressed by regulatory T cells (Tregs). Chemokines are a well-defined group of proteins that were so named due to their chemotactic properties. The current review focuses on key chemokines that not only attract leukocytes but also shape their biological properties. CXCR3 is a chemokine receptor with 3 ligands. We suggest using Ig-based fusion proteins of two of them: CXL9 and CXCL10, to enhance anti-tumor immunity and perhaps transform cold tumors into hot tumors. Potential differences between CXCL9 and CXCL10 regarding ICI are discussed. We also discuss the possibility of targeting the function or deleting a key subset of Tregs that are CCR8+ by monoclonal antibodies to CCR8. These cells are preferentially abundant in several tumors and are likely to be the key drivers in suppressing anti-cancer immune reactivity.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion, P.O. Box 9697, Haifa 31096, Israel
| |
Collapse
|
88
|
van Dorp J, van Montfoort ML, van Dijk N, Hofland I, de Feijter JM, Bergman AM, Hendricksen K, van der Poel HG, van Rhijn BWG, van der Heijden MS. A Serendipitous Preoperative Trial of Combined Ipilimumab Plus Nivolumab for Localized Prostate Cancer. Clin Genitourin Cancer 2021; 20:e173-e179. [PMID: 35016887 DOI: 10.1016/j.clgc.2021.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 11/18/2022]
Affiliation(s)
- Jeroen van Dorp
- Department of Molecular Carcinogenesis, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Maurits L van Montfoort
- Department of Pathology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Nick van Dijk
- Department of Medical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jeantine M de Feijter
- Department of Medical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Andries M Bergman
- Department of Medical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Kees Hendricksen
- Department of Surgical Oncology (Urology), Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Department of Surgical Oncology (Urology), Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Bas W G van Rhijn
- Department of Surgical Oncology (Urology), Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; Department of Urology, Caritas St. Josef Medical Centre, University of Regensburg, Regensburg, Germany
| | - Michiel S van der Heijden
- Department of Molecular Carcinogenesis, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; Department of Medical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| |
Collapse
|
89
|
Sprooten J, Vankerckhoven A, Vanmeerbeek I, Borras DM, Berckmans Y, Wouters R, Laureano RS, Baert T, Boon L, Landolfo C, Testa AC, Fischerova D, Van Holsbeke C, Bourne T, Chiappa V, Froyman W, Schols D, Agostinis P, Timmerman D, Tejpar S, Vergote I, Coosemans A, Garg AD. Peripherally-driven myeloid NFkB and IFN/ISG responses predict malignancy risk, survival, and immunotherapy regime in ovarian cancer. J Immunother Cancer 2021; 9:jitc-2021-003609. [PMID: 34795003 PMCID: PMC8603275 DOI: 10.1136/jitc-2021-003609] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 12/21/2022] Open
Abstract
Background Tumors can influence peripheral immune macroenvironment, thereby creating opportunities for non-invasive serum/plasma immunobiomarkers for immunostratification and immunotherapy designing. However, current approaches for immunobiomarkers’ detection are largely quantitative, which is unreliable for assessing functional peripheral immunodynamics of patients with cancer. Hence, we aimed to design a functional biomarker modality for capturing peripheral immune signaling in patients with cancer for reliable immunostratification. Methods We used a data-driven in silico framework, integrating existing tumor/blood bulk-RNAseq or single-cell (sc)RNAseq datasets of patients with cancer, to inform the design of an innovative serum-screening modality, that is, serum-functional immunodynamic status (sFIS) assay. Next, we pursued proof-of-concept analyses via multiparametric serum profiling of patients with ovarian cancer (OV) with sFIS assay combined with Luminex (cytokines/soluble immune checkpoints), CA125-antigen detection, and whole-blood immune cell counts. Here, sFIS assay’s ability to determine survival benefit or malignancy risk was validated in a discovery (n=32) and/or validation (n=699) patient cohorts. Lastly, we used an orthotopic murine metastatic OV model, with anti-OV therapy selection via in silico drug–target screening and murine serum screening via sFIS assay, to assess suitable in vivo immunotherapy options. Results In silico data-driven framework predicted that peripheral immunodynamics of patients with cancer might be best captured via analyzing myeloid nuclear factor kappa-light-chain enhancer of activated B cells (NFκB) signaling and interferon-stimulated genes' (ISG) responses. This helped in conceptualization of an ‘in sitro’ (in vitro+in situ) sFIS assay, where human myeloid cells were exposed to patients’ serum in vitro, to assess serum-induced (si)-NFκB or interferon (IFN)/ISG responses (as active signaling reporter activity) within them, thereby ‘mimicking’ patients’ in situ immunodynamic status. Multiparametric serum profiling of patients with OV established that sFIS assay can: decode peripheral immunology (by indicating higher enrichment of si-NFκB over si-IFN/ISG responses), estimate survival trends (si-NFκB or si-IFN/ISG responses associating with negative or positive prognosis, respectively), and coestimate malignancy risk (relative to benign/borderline ovarian lesions). Biologically, we documented dominance of pro-tumorigenic, myeloid si-NFκB responseHIGHsi-IFN/ISG responseLOW inflammation in periphery of patients with OV. Finally, in an orthotopic murine metastatic OV model, sFIS assay predicted the higher capacity of chemo-immunotherapy (paclitaxel–carboplatin plus anti-TNF antibody combination) in achieving a pro-immunogenic peripheral milieu (si-IFN/ISG responseHIGHsi-NFκB responseLOW), which aligned with high antitumor efficacy. Conclusions We established sFIS assay as a novel biomarker resource for serum screening in patients with OV to evaluate peripheral immunodynamics, patient survival trends and malignancy risk, and to design preclinical chemo-immunotherapy strategies.
Collapse
Affiliation(s)
- Jenny Sprooten
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ann Vankerckhoven
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Daniel M Borras
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Yani Berckmans
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium
| | - Roxanne Wouters
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium
| | - Raquel S Laureano
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Thais Baert
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium.,Department of Oncology, Leuven Cancer Institute, Laboratory of Gynaecologic Oncology, KU Leuven, Leuven, Belgium
| | | | - Chiara Landolfo
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Queen Charlotte's and Chelsea Hospital, Imperial College, London, UK.,Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Antonia Carla Testa
- Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Dipartimento Scienze della Vita e Sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Tom Bourne
- Queen Charlotte's and Chelsea Hospital, Imperial College, London, UK
| | | | - Wouter Froyman
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Department of Cellular and Molecular Medicine, Cell Death Research and Therapy Laboratory, KU Leuven, Belgium.,VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Dirk Timmerman
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Sabine Tejpar
- Laboratory for Molecular Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ignace Vergote
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium.,Department of Oncology, Leuven Cancer Institute, Laboratory of Gynaecologic Oncology, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Leuven Cancer Institute, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
90
|
Passiglia F, Bertaglia V, Reale ML, Delcuratolo MD, Tabbò F, Olmetto E, Capelletto E, Bironzo P, Novello S. Major breakthroughs in lung cancer adjuvant treatment: Looking beyond the horizon. Cancer Treat Rev 2021; 101:102308. [PMID: 34757306 DOI: 10.1016/j.ctrv.2021.102308] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/15/2022]
Abstract
We are witnessing a silent revolution in the treatment of early stage non-small cell lung cancer (NSCLC), with a series of practice-changing clinical trials enriching the therapeutic perspectives of lung cancer patients with potentially curable disease. The ADAURA study marked the advent of precision medicine and biomarker testing to the early stages setting. The IMPower-010 trial interrupted the negative trend of adjuvant lung cancer immunotherapy, paving the way to the application of immune-checkpoint inhibition in the resected disease. The ITACA trial definitively established no role for tailored adjuvant chemotherapy in NSCLC, while the Lung Art data questioned the efficacy of post-operative radiotherapy for pN2 resected disease. Growing evidence is supporting MRD as effective adjuvant prognostic biomarker to stratify disease's recurrence risk after radical interventions and select best candidates to the adjuvant strategies. This work summarizes the recent major breakthroughs in lung cancer adjuvant treatment, and provides a snapshot of the current real-world scenario, discussing the upcoming challenges and opportunities featuring the clinical management of early stage NSCLC patients.
Collapse
Affiliation(s)
- Francesco Passiglia
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy.
| | - Valentina Bertaglia
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy
| | - Maria Lucia Reale
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy.
| | | | - Fabrizio Tabbò
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy
| | - Emanuela Olmetto
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy
| | - Enrica Capelletto
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy.
| | - Paolo Bironzo
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy.
| | - Silvia Novello
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy.
| |
Collapse
|
91
|
Cancer bio-immunotherapy XVII annual NIBIT (Italian Network for Tumor Biotherapy) meeting, October 11-13 2019, Verona, Italy. Cancer Immunol Immunother 2021; 71:1777-1786. [PMID: 34755203 PMCID: PMC8577637 DOI: 10.1007/s00262-021-03104-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/29/2021] [Indexed: 11/09/2022]
|
92
|
Lee JM, Kim AW, Marjanski T, Falcoz PE, Tsuboi M, Wu YL, Sun SW, Gitlitz BJ. Important Surgical and Clinical End Points in Neoadjuvant Immunotherapy Trials in Resectable NSCLC. JTO Clin Res Rep 2021; 2:100221. [PMID: 34746882 PMCID: PMC8552106 DOI: 10.1016/j.jtocrr.2021.100221] [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: 06/01/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/25/2022] Open
Abstract
Neoadjuvant immunotherapy may improve outcomes in patients with resectable NSCLC and is being evaluated in phase 2 and 3 studies. Nevertheless, preoperative treatment postpones resection; the potential for increased surgical complexity and greater intra- and postoperative morbidity and mortality is an additional consideration. In studies primarily designed to evaluate efficacy, the impact of neoadjuvant immunotherapy on surgery is based on parameters that are poorly defined and reported differently between studies. Defining and reporting common end points among trials would improve understanding and facilitate cross-comparison of different immunotherapy regimens and may facilitate wider adoption of induction therapies by surgeons and oncologists. We propose several surgical end points and related metrics for neoadjuvant immunotherapy in resectable NSCLC. These include the periods from screening to treatment initiation and from last neoadjuvant dose to surgery; reporting of the allowable window for surgery to preclude masking delays caused by induction treatment-related toxicity; complete resection (R0) rate; preoperative downstaging; a standardized list of immune-related adverse events and associated delay to surgery; preoperative attrition; postoperative attrition before adjuvant therapy; and postoperative 30- and 90-day mortality and morbidity rates. Intraoperative end points (blood loss, duration, and type of surgery) and our proposed system of grading complexity based on lymphadenopathy and fibrosis would allow quantitation of technical difficulty and quality of oncologic resection. In conclusion, the standardization, reporting, and prospective inclusion of these end points in study protocols would provide a comparative overview of the impact of different neoadjuvant immunotherapy regimens on surgery and ultimately clinical oncologic outcomes in resectable NSCLC.
Collapse
Affiliation(s)
- Jay M Lee
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Anthony W Kim
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Tomasz Marjanski
- Thoracic Surgery Department, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Masahiro Tsuboi
- Division of Thoracic Surgery, Department of Thoracic Surgery & Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangdong, People's Republic of China
| | - Shawn W Sun
- Product Development Clinical Oncology, Genentech, Inc., South San Francisco, California
| | - Barbara J Gitlitz
- Product Development Clinical Oncology, Genentech, Inc., South San Francisco, California
| |
Collapse
|
93
|
André T, Cohen R. Immune checkpoint inhibitors in colorectal cancer: dream and reality. Lancet Gastroenterol Hepatol 2021; 7:4-6. [PMID: 34688372 DOI: 10.1016/s2468-1253(21)00375-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Thierry André
- Sorbonne University, Saint-Antoine Hospital, AP-HP, and INSERM, Unité Mixte de Recherche Scientifique 938, 75012 Paris, France.
| | - Romain Cohen
- Sorbonne University, Saint-Antoine Hospital, AP-HP, and INSERM, Unité Mixte de Recherche Scientifique 938, 75012 Paris, France
| |
Collapse
|
94
|
Neoadjuvant talimogene laherparepvec plus surgery versus surgery alone for resectable stage IIIB-IVM1a melanoma: a randomized, open-label, phase 2 trial. Nat Med 2021; 27:1789-1796. [PMID: 34608333 DOI: 10.1038/s41591-021-01510-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022]
Abstract
Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1-based intralesional oncolytic immunotherapy approved for the treatment of unresectable melanoma. The present, ongoing study aimed to estimate the treatment effect of neoadjuvant T-VEC on recurrence-free survival (RFS) of patients with advanced resectable melanoma. An open-label, phase 2 trial (NCT02211131) was conducted in 150 patients with resectable stage IIIB-IVM1a melanoma who were randomized to receive T-VEC followed by surgery (arm 1, n = 76) or surgery alone (arm 2, n = 74). The primary endpoint was a 2-year RFS in the intention-to-treat population. Secondary and exploratory endpoints included overall survival (OS), pathological complete response (pCR), safety and biomarker analyses. The 2-year RFS was 29.5% in arm 1 and 16.5% in arm 2 (overall hazard ratio (HR) = 0.75, 80% confidence interval (CI) = 0.58-0.96). The 2-year OS was 88.9% for arm 1 and 77.4% for arm 2 (overall HR = 0.49, 80% CI = 0.30-0.79). The RFS and OS differences between arms persisted at 3 years. In arm 1, 17.1% achieved a pCR. Increased CD8+ density correlated with clinical outcomes in an exploratory analysis. Arm 1 adverse events were consistent with previous reports for T-VEC. The present study met its primary endpoint and estimated a 25% reduction in the risk of disease recurrence for neoadjuvant T-VEC plus surgery versus upfront surgery for patients with resectable stage IIIB-IVM1a melanoma.
Collapse
|
95
|
Beets GL. Colorectal cancer immunotherapy: a treatment quantum leap. Br J Surg 2021; 108:877-878. [PMID: 34378019 DOI: 10.1093/bjs/znab170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022]
Affiliation(s)
- G L Beets
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
96
|
Su YY, Li CC, Lin YJ, Hsu C. Adjuvant versus Neoadjuvant Immunotherapy for Hepatocellular Carcinoma: Clinical and Immunologic Perspectives. Semin Liver Dis 2021; 41:263-276. [PMID: 34130338 DOI: 10.1055/s-0041-1730949] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advancement in systemic therapy, particularly immune checkpoint inhibitor (ICI)-based combination regimens, has transformed the treatment landscape for patients with advanced hepatocellular carcinoma (HCC). The advancement in systemic therapy also provides new opportunities of reducing recurrence after curative therapy through adjuvant therapy or improving resectability through neoadjuvant therapy. Improved recurrence-free survival by adjuvant or neoadjuvant ICI-based therapy has been reported in other cancer types. In this article, developments of systemic therapy in adjuvant and neoadjuvant settings for HCC were reviewed. The design of adjuvant and neoadjuvant therapy using ICI-based regimens and potential challenges of trial conduct and result analysis was discussed. Results from these trials may extend the therapeutic benefit of ICI-based systemic therapy beyond the advanced-stage disease and lead to a new era of multidisciplinary management for HCC.
Collapse
Affiliation(s)
- Yung-Yeh Su
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Chen Li
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yih-Jyh Lin
- Division of General and Transplant Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Liver Cancer Collaborative Oncology Group, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
97
|
Lee JM, Tsuboi M, Brunelli A. Surgical perspective on neoadjuvant immunotherapy in non-small cell lung cancer. Ann Thorac Surg 2021; 114:1505-1515. [PMID: 34339672 DOI: 10.1016/j.athoracsur.2021.06.069] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/25/2021] [Accepted: 06/22/2021] [Indexed: 11/01/2022]
Abstract
BACKGROUND With a 5% improvement in 5-year overall survival achieved with current neoadjuvant or adjuvant chemotherapy, new treatments for resectable non-small cell lung cancer (NSCLC) are urgently needed. The use of immune checkpoint inhibitors (ICI) is established in metastatic NSCLC and is being evaluated in resectable NSCLC. METHODS Publications and conference databases and clinicaltrials.gov were searched for reports on clinical studies of neoadjuvant immunotherapy in patients with early resectable NSCLC. RESULTS Potential advantages of neoadjuvant ICI include earlier treatment of micrometastatic disease; activation of a broader, potentially durable immune response by the whole tumor and associated lymph nodes; and pathologic assessment of neoadjuvant treatment response, which may guide adjuvant therapy. Surgical considerations include delays to surgery, potential disease progression preventing curative resection, and perioperative morbidity and mortality. Surrogate endpoints of efficacy (pathologic complete response, major pathologic response) and biomarkers predictive of outcome (programmed death ligand 1 expression, tumor mutational burden and circulating tumor DNA) can accelerate clinical trial completion and early-stage treatment development; their application in neoadjuvant ICI studies in NSCLC is reviewed. CONCLUSIONS Phase 2 trials of neoadjuvant ICI alone or with chemotherapy showed encouraging safety and efficacy in patients with resectable NSCLC, warranting the ongoing phase 3 studies of neoadjuvant immunotherapy plus chemotherapy. Preoperative and intraoperative unresectability following neoadjuvant ICI appear comparable to neoadjuvant chemotherapy. To help thoracic surgeons and medical oncologists to distinguish amongst ICI beyond efficacy as phase 3 data emerge, surgery-related endpoints for perioperative morbidity, mortality, and complexity should be defined, standardized, incorporated into trial designs, and reported.
Collapse
Affiliation(s)
- Jay M Lee
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA, Division of Thoracic Surgery.
| | - Masahiro Tsuboi
- National Cancer Center Hospital East, Kashiwanoha, Kashiwa, Chiba, Japan Division of Thoracic Surgery, Department of Thoracic Oncology
| | - Alessandro Brunelli
- University of Leeds and St. James's University Hospital, Leeds, UK, Department of Thoracic Surgery
| |
Collapse
|
98
|
Paijens ST, Vledder A, Loiero D, Duiker EW, Bart J, Hendriks AM, Jalving M, Workel HH, Hollema H, Werner N, Plat A, Wisman GBA, Yigit R, Arts H, Kruse AJ, de Lange N, Koelzer VH, de Bruyn M, Nijman HW. Prognostic image-based quantification of CD8CD103 T cell subsets in high-grade serous ovarian cancer patients. Oncoimmunology 2021; 10:1935104. [PMID: 34123576 PMCID: PMC8183551 DOI: 10.1080/2162402x.2021.1935104] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/20/2021] [Indexed: 11/06/2022] Open
Abstract
CD103-positive tissue resident memory-like CD8+ T cells (CD8CD103 TRM) are associated with improved prognosis across malignancies, including high-grade serous ovarian cancer (HGSOC). However, whether quantification of CD8, CD103 or both is required to improve existing survival prediction and whether all HGSOC patients or only specific subgroups of patients benefit from infiltration, remains unclear. To address this question, we applied image-based quantification of CD8 and CD103 multiplex immunohistochemistry in the intratumoral and stromal compartments of 268 advanced-stage HGSOC patients from two independent clinical institutions. Infiltration of CD8CD103 immune cell subsets was independent of clinicopathological factors. Our results suggest CD8CD103 TRM quantification as a superior method for prognostication compared to single CD8 or CD103 quantification. A survival benefit of CD8CD103 TRM was observed only in patients treated with primary cytoreductive surgery. Moreover, survival benefit in this group was limited to patients with no macroscopic tumor lesions after surgery. This approach provides novel insights into prognostic stratification of HGSOC patients and may contribute to personalized treatment strategies in the future.
Collapse
Affiliation(s)
- S. T. Paijens
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. Vledder
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - D. Loiero
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - E. W. Duiker
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J. Bart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. M. Hendriks
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M. Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. H. Workel
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. Hollema
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - N. Werner
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. Plat
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G. B. A. Wisman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - R. Yigit
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. Arts
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. J. Kruse
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - N.M. de Lange
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - V. H. Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - M. de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. W. Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
99
|
Early memory differentiation and cell death resistance in T cells predicts melanoma response to sequential anti-CTLA4 and anti-PD1 immunotherapy. Genes Immun 2021; 22:108-119. [PMID: 34079092 DOI: 10.1038/s41435-021-00138-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/11/2021] [Accepted: 05/21/2021] [Indexed: 11/09/2022]
Abstract
Immune checkpoint blockers (ICBs)-based immunotherapy has revolutionised oncology. However, the benefits of ICBs are limited to only a subset of patients. Herein, the biomarkers-driven application of ICBs promises to increase their efficacy. Such biomarkers include lymphocytic IFNγ-signalling and/or cytolytic activity (granzymes and perforin-1) footprints, whose levels in pre-treatment tumours can predict favourable patient survival following ICB-treatment. However, it is not clear whether such biomarkers have the same value in predicting survival of patients receiving first-line anti-CTLA4 ICB-therapy, and subsequently anti-PD1 ICB-therapy (i.e., sequential ICB-immunotherapy regimen). To address this, we applied highly integrated systems/computational immunology approaches to existing melanoma bulk-tumour transcriptomic and single-cell (sc)RNAseq data originating from immuno-oncology clinical studies applying ICB-treatment. Interestingly, we observed that CD8+/CD4+T cell-associated IFNγ-signalling or cytolytic activity signatures fail to predict tumour response in patients treated with anti-CTLA4 ICB-therapy as a first-line and anti-PD1 ICB-therapy in the second-line setting. On the contrary, signatures associated with early memory CD8+/CD4+T cells (integrating TCF1-driven stem-like transcriptional programme), capable of resisting cell death/apoptosis, better predicted objective response rates to ICB-immunotherapy, and favourable survival in the setting of sequential ICB-immunotherapy. These observations suggest that sequencing of ICB-therapy might have a specific impact on the T cell-repertoire and may influence the predictive value of tumoural immune biomarkers.
Collapse
|
100
|
Nakano K. Progress of molecular targeted therapy for head and neck cancer in clinical aspects. MOLECULAR BIOMEDICINE 2021; 2:15. [PMID: 35006440 PMCID: PMC8607362 DOI: 10.1186/s43556-021-00032-5] [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: 10/27/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
Since the body's head and neck area affects many functions such as breathing, swallowing, and speaking, systemic treatments to head and neck cancer patients are important not only for survival but also for preserving functions and quality of life. With the progress that has been made in molecular targeted therapy, anti-EGFR antibody (cetuximab) and immune checkpoint inhibitors (nivolumab, pembrolizumab) have provided survival benefits to head and neck cancer patients and are approved for clinical practice. Clinical trials incorporating these new drugs for patients with locally advanced head/neck cancers are underway. However, the existing clinical evidence regarding molecular targeted drugs for head and neck cancers is based mostly on clinical trials allocated to squamous cell carcinoma patients. New targeted therapies for non-squamous cell carcinoma patients were recently reported, e.g., tyrosine kinase inhibitors for the treatment of thyroid cancers and HER2-targeted therapy for salivary gland cancers. With the goal of improving local control, molecular targeted treatment strategies as salvage local therapy are being investigated, including boron neutron capture therapy (BNCT) and near-infrared photoimmunotherapy (NIR-PIT). Herein the history and landscape of molecular targeted therapy for head and neck cancers are summarized and reviewed.
Collapse
Affiliation(s)
- Kenji Nakano
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, 135-8550, Japan.
| |
Collapse
|