1
|
Nesbitt C, Van Der Walt A, Butzkueven H, Devitt B, Jokubaitis VG. Multiple sclerosis and cancer: Navigating a dual diagnosis. Mult Scler 2024; 30:1714-1736. [PMID: 39347791 DOI: 10.1177/13524585241274523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Healthcare breakthroughs are extending the lives of multiple sclerosis (MS) patients and cancer survivors, creating a growing cohort of individuals navigating a dual diagnosis. Determining the relationship between MS and cancer risk remains challenging, with inconclusive findings confounded by age, risk exposures, comorbidities, genetics and the ongoing introduction of new MS disease-modifying therapies (DMTs) across study periods.This research places significant emphasis on cancer survival, with less attention given to the impact on MS outcomes. Our review explores the existing literature on MS, cancer risk and the intersection of DMTs and cancer treatments. We aim to navigate the complexities of managing MS in cancer survivors to optimise outcomes for both conditions. Continuous research and the formulation of treatment guidelines are essential for guiding future care. Collaboration between neuro-immunology and oncology is crucial, with a need to establish databases for retrospective and ultimately prospective analysis of outcomes in these rapidly evolving fields.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Bianca Devitt
- Department of Oncology, Eastern Health Clinical School, Monash University, Melbourne, VIC, Australia
- Oncology Clinical Trials Unit, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Nicolas E, Kosmider B, Cukierman E, Borghaei H, Golemis EA, Borriello L. Cancer treatments as paradoxical catalysts of tumor awakening in the lung. Cancer Metastasis Rev 2024; 43:1165-1183. [PMID: 38963567 PMCID: PMC11554904 DOI: 10.1007/s10555-024-10196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Much of the fatality of tumors is linked to the growth of metastases, which can emerge months to years after apparently successful treatment of primary tumors. Metastases arise from disseminated tumor cells (DTCs), which disperse through the body in a dormant state to seed distant sites. While some DTCs lodge in pre-metastatic niches (PMNs) and rapidly develop into metastases, other DTCs settle in distinct microenvironments that maintain them in a dormant state. Subsequent awakening, induced by changes in the microenvironment of the DTC, causes outgrowth of metastases. Hence, there has been extensive investigation of the factors causing survival and subsequent awakening of DTCs, with the goal of disrupting these processes to decrease cancer lethality. We here provide a detailed overview of recent developments in understanding of the factors controlling dormancy and awakening in the lung, a common site of metastasis for many solid tumors. These factors include dynamic interactions between DTCs and diverse epithelial, mesenchymal, and immune cell populations resident in the lung. Paradoxically, among key triggers for metastatic outgrowth, lung tissue remodeling arising from damage induced by the treatment of primary tumors play a significant role. In addition, growing evidence emphasizes roles for inflammation and aging in opposing the factors that maintain dormancy. Finally, we discuss strategies being developed or employed to reduce the risk of metastatic recurrence.
Collapse
Affiliation(s)
- Emmanuelle Nicolas
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Beata Kosmider
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Hossein Borghaei
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Lucia Borriello
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA.
| |
Collapse
|
3
|
Misawa K, Bhat H, Adusumilli PS, Hou Z. Combinational CAR T-cell therapy for solid tumors: Requisites, rationales, and trials. Pharmacol Ther 2024; 266:108763. [PMID: 39617146 DOI: 10.1016/j.pharmthera.2024.108763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved potent antitumor efficacy in hematological malignancies; however, because of limitations in CAR T-cell recruitment, infiltration, activation, and functional persistence in the tumor, its efficacy in solid tumors has been suboptimal. To overcome these challenges, combinational strategies that include chemotherapy, radiation therapy, or immune checkpoint inhibitor agent therapy with CAR T-cell therapy are being investigated. The established functional characteristics of the abovementioned therapies provide a rationale for the use of a combinational approach with CAR T cells. Chemotherapy reshapes the peritumoral stroma, decreases the immunosuppressive cell population, and promotes a proinflammatory milieu, all of which allow for increased recruitment, infiltration, and accumulation of CAR T cells. Radiation therapy promotes a chemokine gradient, which augments tumor infiltration by CAR T cells and further increases expression of tumor-associated antigens, allowing for increased activation of CAR T cells. Immune checkpoint inhibitor agent therapy inactivates T-cell exhaustion pathways-most notably, the PD1/PDL1 pathway-thereby improving the functional persistence of CAR T cells and promoting endogenous immunity. In this review, we discuss the requisites and rationales for combinational therapy, and we review 25 ongoing phase I and II clinical trials, of which 4 use chemotherapy, 3 use radiation therapy, 11 use immunotherapy, and 7 use another agent. While safety, efficacy, and improved outcomes are the primary goals of these ongoing studies, the knowledge gained from them will help pave the way for subsequent studies focused on optimizing combinational regimens and identifying predictive biomarkers.
Collapse
Affiliation(s)
- Kyohei Misawa
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Hina Bhat
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
4
|
Sjoerdsma JN, Bromley EK, Shin J, Hilliard T, Liu Y, Horgan C, Hwang G, Bektas M, Omstead D, Kiziltepe T, Stack MS, Bilgicer B. Combination non-targeted and sGRP78-targeted nanoparticle drug delivery outperforms either component to treat metastatic ovarian cancer. J Control Release 2024; 375:438-453. [PMID: 39271060 PMCID: PMC11486564 DOI: 10.1016/j.jconrel.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Metastatic ovarian cancer (MOC) is highly deadly, due in part to the limited efficacy of standard-of-care chemotherapies to metastatic tumors and non-adherent cancer cells. Here, we demonstrated the effectiveness of a combination therapy of GRP78-targeted (TNPGRP78pep) and non-targeted (NP) nanoparticles to deliver a novel DM1-prodrug to MOC in a syngeneic mouse model. Cell surface-GRP78 is overexpressed in MOC, making GRP78 an optimal target for selective delivery of nanoparticles to MOC. The NP + TNPGRP78pep combination treatment reduced tumor burden by 15-fold, compared to untreated control. Increased T cell and macrophage levels in treated groups also suggested antitumor immune system involvement. The NP and TNPGRP78pep components functioned synergistically through two proposed mechanisms of action. The TNPGRP78pep targeted non-adherent cancer cells in the peritoneal cavity, preventing the formation of new solid tumors, while the NP passively targeted existing solid tumor sites, providing a sustained release of the drug to the tumor microenvironment.
Collapse
Affiliation(s)
- Jenna N Sjoerdsma
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily K Bromley
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jaeho Shin
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Tyvette Hilliard
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yueying Liu
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Caitlin Horgan
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Gyoyeon Hwang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael Bektas
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - David Omstead
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Tanyel Kiziltepe
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - M Sharon Stack
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Basar Bilgicer
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Center for Rare & Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
5
|
Lok V, Olson-McPeek S, Spiegelhoff G, Cortez J, Detz D, Czerniecki B. Immunotherapies in breast cancer: harnessing the cancer immunity cycle. Expert Opin Ther Targets 2024; 28:925-935. [PMID: 39523444 DOI: 10.1080/14728222.2024.2427038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Immunotherapies have found limited success in breast cancerdue to significant challenges within the tumor that block T-cell activity and function. AREAS COVERED The current review discusses clinically relevant immunotherapeutics and trials within the framework of the cancer-immunity cycle. EXPERT OPINION Current therapies such as antibody-drug conjugates and immune checkpoint blockade require proper biomarker selection, such as PD1 expression and the degree of tumor-infiltrating lymphocyte (TIL) infiltration to subset potential responders. HER2 and other tumor-associated antigens have served as valuable benchmarks for developing novel therapies, such as antibody engagers and CAR T-cells. However, further research is essential to identify and validate new target antigens that can enhance therapeutic efficacy and broaden the clinical applicability of these approaches.
Collapse
Affiliation(s)
- Vincent Lok
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - Sy Olson-McPeek
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - Grace Spiegelhoff
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - Jaqueline Cortez
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - David Detz
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - Brian Czerniecki
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
6
|
Mackenzie NJ, Zimmermann K, Nicholls C, Perera MP, Ngoo A, Jeffery PL, Vela I, Kenna TJ, Williams ED, Thomas PB. Altered immunophenotypic expression in the peripheral bladder cancer immune landscape. Immunol Cell Biol 2024; 102:949-962. [PMID: 39474781 DOI: 10.1111/imcb.12829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 11/09/2024]
Abstract
Treatments targeting the immune system only benefit a subset of patients with bladder cancer (BC). Biomarkers predictive of BC progression and response to specific therapeutic interventions are required. We evaluated whether peripheral blood immune subsets and expression of clinically relevant immune checkpoint markers are associated with clinicopathologic features of BC. Peripheral blood mononuclear cells isolated from blood collected from 23 patients with BC and 9 age-matched unaffected-by-cancer control donors were assessed using a 21-parameter flow cytometry panel composed of markers of T, B, natural killer and myeloid populations and immune checkpoint markers. Patients with BC had significantly lower numbers of circulating CD19+ B cells and elevated circulating CD4+CD8+ T cells compared with the control cohort. Immune checkpoint markers programmed cell death protein 1 (PD-1) and T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) were elevated in the total peripheral immune cell population in patients with BC. Within the BC cohort, PD-1 expression in T and myeloid cells was elevated in muscle-invasive compared with non-muscle-invasive disease. In addition, elevated T, B and myeloid PD-1 cell surface expression was significantly associated with tumor stage, suggesting that measures of peripheral immune cell exhaustion may be a predictor of tumor progression in BC. Finally, positive correlations between expression levels of the various immune checkpoints both overall and within key peripheral blood immune subsets collected from patients with BC were observed, highlighting likely coregulation of peripheral immune checkpoint expression. The peripheral blood immunophenotype in patients with BC is altered compared with cancer-free individuals. Understanding this dysregulated immune profile will contribute to the identification of diagnostic and prognostic indicators to guide effective immune-targeted, personalized treatments.
Collapse
Affiliation(s)
- Nathan J Mackenzie
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Kate Zimmermann
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Centre for Immunology and Infection Control, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Microbiome Research, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Clarissa Nicholls
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Mahasha Pj Perera
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Alexander Ngoo
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Penny L Jeffery
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Ian Vela
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Tony J Kenna
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Centre for Immunology and Infection Control, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Microbiome Research, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Elizabeth D Williams
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Patrick B Thomas
- Queensland University of Technology (QUT), School of Biomedical Sciences at Translational Research Institute (TRI), Brisbane, QLD, Australia
- Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
7
|
Michal R, Andrea W, Jana H, Lucie P, Eva R, Lubos B, Kateřina K, Iva H, Igor K. The impact of supervised and home exercise activity intervention on circulating immune cell numbers in cancer patients. Heliyon 2024; 10:e39320. [PMID: 39640787 PMCID: PMC11620229 DOI: 10.1016/j.heliyon.2024.e39320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Malignant diseases challenge clinicians to balance treatment intensity and patient quality of life. Regular physical activity positively impacts mental and physical health, benefiting sleep patterns, heart rate, and overall health. Moreover, telehealth physical exercise training represents a viable option for maintaining intrinsic capacity. The American Cancer Society highlights exercise's role in helping patients cope with anti-cancer treatment side effects. In the Czech Republic, there is no fitness-promoting protocol for cancer patients, despite recognized benefits. Exercise may also enhance immune function, with moderate-intensity exercise potentially positively affecting immune cell counts. Objective This study aimed to analyze the long-term effects of exercise on circulating immune cells in patients undergoing treatment for solid malignancies. Patients and methods 49 participants were recruited at the Masaryk Memorial Cancer Institute, Czech Republic, starting September 2021. Participants were randomized into an experimental (SAPA = undergoing monitored exercise program) group (N = 16; madian age: 46,6; median BMI: 25,1) and a control (CO) group (N = 33; madian age: 52,0; median BMI: 25,3). Flow cytometry was used to examine cellular immunological profiles. The exercise program involved thrice-weekly sessions conducted online. Results SAPA group showed stable lymphocyte counts post-exercise (percentage chance: +0,5 %; p = 0,256; effect size r = -0,284), while the CO group exhibited a significant drop (percentage chance: -23,0 %; p = 0,015; effect size r = -0,423). B lymphocyte numbers were significantly higher in the SAPA group post-exercise compared to the CO group (p = 0,003; effect size r = -0,422). The number of Th-lymphocytes, T-c lymphocytes, T-gamma/delta lymphocytes, and NK cells remained stable in SAPA but dropped in CO group. Conclusion Exercise's impact on the immune system in cancer patients shows promise, with differences noted between acute and chronic exercise effects. Previous studies on acute exercise indicate a rise in immune cell counts, supporting our findings of stable or increased immune cells with controlled exercise in cancer patients. Controlled physical activity stabilizes or increases certain immune cell populations in patients undergoing chemotherapy for solid malignancies, highlighting the potential benefits of incorporating exercise into cancer treatment protocols.
Collapse
Affiliation(s)
- Rihacek Michal
- Department of Hematology and Transfusion, AGEL Central Moravia Hospitals, AGEL Prostejov Hospital, Mathonova 1, 796 01, Prostejov, Czechia
- Department of Laboratory Medicine, AGEL Central Moravia Hospitals, Mathonova 1, 796 01, Prostejov, Czechia
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czechia
| | - Wagnerova Andrea
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czechia
- Department of Laboratory Medicine, University Hospital Brno, Jihlavska 20, 625 00, Brno, Czechia
| | - Halamkova Jana
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czechia
| | - Pehalova Lucie
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Kamenice 126/3, 62500, Brno, Czechia
- Institute of Health Information and Statistics of the Czech Republic, Palackeho namesti 4, 12801 Prague 2, Czechia
| | - Rihackova Eva
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavska 20, 625 00, Brno, Czechia
| | - Boucek Lubos
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czechia
| | - Kapounková Kateřina
- Department of Physical Activities and Health Sciences, Faculty of Sports, Masaryk University, Kamenice 5, 625 00, Brno, Czechia
| | - Hrnčiříková Iva
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czechia
| | - Kiss Igor
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53, Brno, Czechia
| |
Collapse
|
8
|
Oliveira J, Cruz P, Dias TR, Sousa-Pimenta M, Almeida B, Soares B, Sousa H, Costa R, Ochoa C, Dias F, Medeiros R. Humoral Response to SARS-CoV-2 Vaccine-Boost in Cancer Patients: A Case Series from a Southern European Cancer Center. Vaccines (Basel) 2024; 12:1207. [PMID: 39591110 PMCID: PMC11598862 DOI: 10.3390/vaccines12111207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Cancer patients face a greater risk of complications and death after contracting the SARS-CoV-2 virus. Booster doses of the COVID-19 vaccine were suggested to provide additional protection. This study aimed to assess how cancer patients' immune systems respond to the booster shots and categorize their responses. METHODS We analyzed 735 samples from 422 individuals, including patients followed at the Portuguese Oncology Institute of Porto (IPO-Porto). Three cohorts were recruited, and blood samples were collected 3- and 6-months post-booster dose: cohort 1 cancer patients (also collected before the booster); cohort 2 cancer patients; and cohort 3 (healthy individuals). Humoral immune response was evaluated by analyzing IgG levels against the SARS-CoV-2 Spike (S) protein. IgG levels against the SARS-CoV-2 Nucleocapsid(N) protein was also analyzed in order to address previous contact with the virus. RESULTS Among Cohort 1 patients with solid tumors, when compared to pre-boost, IgG S levels increased 3 months after the boost and remained high after 6 months. Patients with hematologic tumors demonstrated lower IgG S levels at both timepoints. Comparing the IgG S levels among hematological tumors, solid tumors, and healthy individuals in both timepoints we observed that the healthy individuals had the strongest IgG S response, followed by the solid, and, lastly, the hematologic tumors. Solid tumor patients undergoing chemotherapy had reduced IgG S levels, especially those on high febrile neutropenia risk regimens. CONCLUSIONS In conclusion, cancer patients have a weaker immune response to the SARS-CoV-2 vaccine, especially those with hematological cancers. Chemotherapy and febrile neutropenia risk further reduce booster effectiveness. Further research is needed to optimize vaccine timing for cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Júlio Oliveira
- Department of Medical Oncology, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (J.O.); (P.C.); (B.S.)
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Pedro Cruz
- Department of Medical Oncology, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (J.O.); (P.C.); (B.S.)
| | - Tânia R. Dias
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal;
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (B.A.); (H.S.); (F.D.)
| | - Mário Sousa-Pimenta
- Department of Oncohematology, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal;
| | - Beatriz Almeida
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (B.A.); (H.S.); (F.D.)
- Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
| | - Bruno Soares
- Department of Medical Oncology, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (J.O.); (P.C.); (B.S.)
| | - Hugo Sousa
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (B.A.); (H.S.); (F.D.)
- Laboratory Medicine, Clinical Pathology Department, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
| | - Rui Costa
- Department of Occupational Health, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (R.C.); (C.O.)
| | - Carlos Ochoa
- Department of Occupational Health, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (R.C.); (C.O.)
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (B.A.); (H.S.); (F.D.)
| | - Rui Medeiros
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal;
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (B.A.); (H.S.); (F.D.)
- Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
- Laboratory Medicine, Clinical Pathology Department, Portuguese Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- Research Innovation and Development Institute (FP-I3ID), Faculty of Health Sciences of Fernando Pessoa University (UFP), 4249-004 Porto, Portugal
| |
Collapse
|
9
|
Dixon-Douglas J, Virassamy B, Clarke K, Hun M, Luen SJ, Savas P, van Geelen CT, David S, Francis PA, Salgado R, Michiels S, Loi S. Sustained lymphocyte decreases after treatment for early breast cancer. NPJ Breast Cancer 2024; 10:94. [PMID: 39433772 PMCID: PMC11493948 DOI: 10.1038/s41523-024-00698-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
The role of adaptive immunity in long-term outcomes in early breast cancer is increasingly recognised. Standard (neo)adjuvant chemotherapy can have adverse effects on immune cells. We conducted a retrospective longitudinal study of full blood counts (FBC) of 200 patients receiving (neo)adjuvant chemotherapy for early breast cancer at a single institution. FBC results at four time points from pre-treatment to 12 months post-chemotherapy were analysed. Flow cytometry was performed for patients with matched pre- and post-chemotherapy peripheral blood mononuclear cell samples. A significant decrease in absolute lymphocyte count at 12 months post-chemotherapy was observed (p < 0.01), most pronounced in pre-menopausal patients (n = 73; p < 0.01), patients receiving dose-dense chemotherapy regimens (n = 60; p < 0.01) and patients receiving adjuvant radiotherapy (n = 147, p < 0.01). In pre-menopausal patients, significant changes in CD4+ T cells subsets post-chemotherapy were observed. Further investigation, including long-term clinical outcomes, is needed to meaningfully improve long-term anti-tumour immunity.
Collapse
Affiliation(s)
- Julia Dixon-Douglas
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
- Institut Gustave Roussy, INSERM U981, PRISM Center, F-94805, Villejuif, France
| | - Balaji Virassamy
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Kylie Clarke
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael Hun
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Stephen J Luen
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | - Peter Savas
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | | | - Steven David
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Prudence A Francis
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | - Roberto Salgado
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Pathology, ZAS-Hospitals, Antwerp, Belgium
| | - Stefan Michiels
- Institut Gustave Roussy, INSERM U981, PRISM Center, F-94805, Villejuif, France
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia.
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
10
|
Maneechai K, Khopanlert W, Noiperm P, Udomsak P, Viboonjuntra P, Julamanee J. Generation of ex vivo autologous hematopoietic stem cell-derived T lymphocytes for cancer immunotherapy. Heliyon 2024; 10:e38447. [PMID: 39398019 PMCID: PMC11467635 DOI: 10.1016/j.heliyon.2024.e38447] [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: 05/04/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
CD19CAR-T cell therapy demonstrated promising outcomes in relapsed/refractory B-cell malignancies. Nonetheless, the limited T-cell function and ineffective T-cell apheresis for therapeutic purposes are still concern in heavily pretreated patients. We investigated the feasibility of generating hematopoietic stem cell-derived T lymphocytes (HSC-T) for cancer immunotherapy. The patients' autologous peripheral blood HSCs were enriched for CD34+ and CD3+ cells. The CD34+ cells were then cultured following three steps of lymphoid progenitor differentiation, T-cell differentiation, and T-cell maturation processes. HSC-T cells were successfully generated with robust fold expansion of 3735 times. After lymphoid progenitor differentiation, CD5+ and CD7+ cells remarkably increased (65-84 %) while CD34+ cells consequentially declined. The mature CD3+ cells were detected up to 40 % and 90 % on days 42 and 52, respectively. The majority of HSC-T population was naïve phenotype compared to CD3-T cells (73 % vs 34 %) and CD8:CD4 ratio was 2:1. The higher level of cytokine and cytotoxic granule secretion in HSC-T was observed after activation. HSC-T cells were assessed for clinical application and found that CD19CAR-transduced HSC-T cells demonstrated higher cytokine secretion and a trend of superior cytotoxicity against CD19+ target cells compared to control CAR-T cells. A chronic antigen stimulation assay revealed similar T-cell proliferation, stemness, and exhaustion phenotypes among CAR-T cell types. In conclusions, autologous HSC-T was feasible to generate with preserved T-cell efficacy. The HSC-T cells are potentially utilized as an alternative option for cellular immunotherapy.
Collapse
Affiliation(s)
- Kajornkiat Maneechai
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Wannakorn Khopanlert
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Anatomical Pathology Unit, Division of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Panarat Noiperm
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Phakaporn Udomsak
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Pongtep Viboonjuntra
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Jakrawadee Julamanee
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| |
Collapse
|
11
|
Quintana A, Saini KS, Vidal L, Peg V, Slebe F, Loibl S, Curigliano G, Schmid P, Cortes J. Window of opportunity trials with immune checkpoint inhibitors in triple-negative breast cancer. ESMO Open 2024; 9:103713. [PMID: 39357122 PMCID: PMC11480225 DOI: 10.1016/j.esmoop.2024.103713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/08/2024] [Accepted: 08/18/2024] [Indexed: 10/04/2024] Open
Abstract
Patients with triple-negative breast cancer (TNBC) have a relatively poor clinical outcome. The immune checkpoint inhibitor (ICI) pembrolizumab combined with chemotherapy is the current standard of care in TNBC patients with stage II and III. Monotherapy with ICIs has not been comprehensively assessed in the neoadjuvant setting in TNBC patients, given unfavorable results in metastatic trials. ICIs, however, have been tested in the window of opportunity (WOO) before surgery or standard chemotherapy-based neoadjuvant treatment. The WOO design is well suited to assess an ICI alone or in combination with other ICIs, targeted therapy, radiotherapy or cryotherapy, and measure their pharmacodynamic and clinical effect in this treatment-naive population. Some patients show a good response to ICIs in WOO studies. Biomarkers like tumor-infiltrating lymphocytes, programmed death ligand-1, and interferon-γ signature may predict activity and may identify patients likely to benefit from ICIs. Moreover, an increase in tumor-infiltrating lymphocytes, programmed death ligand-1 expression or T cell receptor expansion following administration of ICIs in the WOO setting could potentially inform of immunotherapy benefit, which would allow tailoring further treatment. This article reviews WOO trials that assessed immunotherapy in the early-stage TNBC population, and how these results could be translated to test de-escalation strategies of neoadjuvant chemotherapy and immunotherapy without compromising a patient's prognosis.
Collapse
Affiliation(s)
- A Quintana
- Breast Cancer Unit, Vall d'Hebrón Institute of Oncology, Barcelona, Spain.
| | - K S Saini
- Fortrea, Inc., Durham, USA; Addenbrooke's Hospital, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, UK
| | | | - V Peg
- Biomedical Research Network Centre in Oncology (CIBERONC), Madrid; Department of Pathology, Vall d'Hebron University Hospital, Barcelona; Departament of Medicine, Universitat Autonoma de Barcelona, Barcelona
| | - F Slebe
- Medica Scientia Innovation Research (MedSIR), Barcelona, Spain; Oncoclínicas & Co, Jersey City, New Jersey and Sao Paulo, Brazil
| | - S Loibl
- German Breast Group, GBG Forschungs GmbH, Neu-Isenburg, Germany
| | - G Curigliano
- European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - P Schmid
- Barts Cancer Institute, Queen Mary University London, London, UK
| | - J Cortes
- Medica Scientia Innovation Research (MedSIR), Barcelona, Spain; Oncoclínicas & Co, Jersey City, New Jersey and Sao Paulo, Brazil; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid; International Breast Cancer Center, Pangaea Oncology, Quironsalud Group, Barcelona; IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana, Madrid, Spain
| |
Collapse
|
12
|
Hong W, Zhang L, Qi Y, Wang Y, Wang W. Impact of Chemotherapy on Circulating Lymphocyte Subsets in Lung Cancer Patients. Cancer Manag Res 2024; 16:1205-1213. [PMID: 39282611 PMCID: PMC11401525 DOI: 10.2147/cmar.s475967] [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: 05/18/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Lung cancer remains a leading cause of cancer-related death and chemotherapy stands as a fundamental component in therapy. Chemotherapy-induced myelosuppression encompasses a spectrum of hematological declines, including not only neutrophils but also lymphocytes, hemoglobin levels and platelets. This retrospective cohort study investigates alterations in peripheral blood lymphocyte subsets. By uncovering these changes, our goal is to refine patient management strategies, ensuring that the benefits of chemotherapy are maximized while minimizing its detrimental effects. Patients and Methods We retrospectively analyzed 159 lung cancer patients. Patients were categorized as "NT" (n=108, no previous anti-tumor therapy), and "PT" (n=51, prior therapy followed by at least a two-month treatment-free interval). Post-chemotherapy, patients were reassessed and grouped into "EarlyCycle" for those who underwent four or fewer cycles, and "LateCycle" for those who underwent more than four cycles. Results The study focused on analyzing the percentages of lymphocyte subsets, including T cells (CD4+, CD8+), B cells, and natural killer (NK) cells, across these groups. For T cells, the EarlyCycle group exhibited a significant increase compared to NT (0.7783 vs 0.7271; p=0.0017) and PT (0.7783 vs 0.6804; p=1.6e-05). B cells showed a significant decrease from NT to LateCycle (0.1014 vs 0.0817; p=2.2e-05) and from PT to LateCycle (0.1317 vs 0.0817; p=6.2e-10). NK cells significantly decreased in the EarlyCycle group compared to NT (0.1109 vs 0.1462; p=0.00816) and PT (0.1109 vs 0.1513; p=0.00992), with no significant change in the LateCycle group compared to either NT or PT (p>0.05). Conclusion Chemotherapy significantly affects lymphocyte subsets in a treatment-specific manner. The EarlyCycle group experienced a reduction in NK cell and an increase in T cell, suggesting a damage of innate immunity and an early shift towards adaptive immunity. The LateCycle group showed a substantial decrease in B cell, indicating a delayed effect on humoral immunity components.
Collapse
Affiliation(s)
- Wei Hong
- Oncology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Lei Zhang
- Oncology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Youkun Qi
- Pharmacy, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Yanjun Wang
- Oncology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Wentao Wang
- Critical Care Medicine, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| |
Collapse
|
13
|
Bose B, Bozdag S. Identifying cell lines across pan-cancer to be used in preclinical research as a proxy for patient tumor samples. Commun Biol 2024; 7:1101. [PMID: 39244634 PMCID: PMC11380668 DOI: 10.1038/s42003-024-06812-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
In pre-clinical trials of anti-cancer drugs, cell lines are utilized as a model for patient tumor samples to understand the response of drugs. However, in vitro culture of cell lines, in general, alters the biology of the cell lines and likely gives rise to systematic differences from the tumor samples' genomic profiles; hence the drug response of cell lines may deviate from actual patients' drug response. In this study, we computed a similarity score for the selection of cell lines depicting the close and far resemblance to patient tumor samples in twenty-two different cancer types at genetic, genomic, and epigenetic levels integrating multi-omics datasets. We also considered the presence of immune cells in tumor samples and cancer-related biological pathways in this score which aids personalized medicine research in cancer. We showed that based on these similarity scores, cell lines were able to recapitulate the drug response of patient tumor samples for several FDA-approved cancer drugs in multiple cancer types. Based on these scores, several of the high-rank cell lines were shown to have a close likeness to the corresponding tumor type in previously reported in vitro experiments.
Collapse
Affiliation(s)
- Banabithi Bose
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, USA.
- Department of Mathematics, University of North Texas, Denton, TX, USA.
- BioDiscovery Institute, University of North Texas, Denton, TX, USA.
- Center for Computational Life Sciences, University of North Texas, Denton, TX, USA.
| |
Collapse
|
14
|
Kovale L, Singh MK, Kim J, Ha J. Role of Autophagy and AMPK in Cancer Stem Cells: Therapeutic Opportunities and Obstacles in Cancer. Int J Mol Sci 2024; 25:8647. [PMID: 39201332 PMCID: PMC11354724 DOI: 10.3390/ijms25168647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer stem cells represent a resilient subset within the tumor microenvironment capable of differentiation, regeneration, and resistance to chemotherapeutic agents, often using dormancy as a shield. Their unique properties, including drug resistance and metastatic potential, pose challenges for effective targeting. These cells exploit certain metabolic processes for their maintenance and survival. One of these processes is autophagy, which generally helps in energy homeostasis but when hijacked by CSCs can help maintain their stemness. Thus, it is often referred as an Achilles heel in CSCs, as certain cancers tend to depend on autophagy for survival. Autophagy, while crucial for maintaining stemness in cancer stem cells (CSCs), can also serve as a vulnerability in certain contexts, making it a complex target for therapy. Regulators of autophagy like AMPK (5' adenosine monophosphate-activated protein kinase) also play a crucial role in maintaining CSCs stemness by helping CSCs in metabolic reprogramming in harsh environments. The purpose of this review is to elucidate the interplay between autophagy and AMPK in CSCs, highlighting the challenges in targeting autophagy and discussing therapeutic strategies to overcome these limitations. This review focuses on previous research on autophagy and its regulators in cancer biology, particularly in CSCs, addresses the remaining unanswered questions, and potential targets for therapy are also brought to attention.
Collapse
Affiliation(s)
- Lochana Kovale
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| |
Collapse
|
15
|
Suárez GM, Catalá M, Peña Y, Portela S, Añé-Kourí AL, González A, Lorenzo-Luaces P, Díaz M, Molina MDLA, Pereira K, Hernández JDLC, Reyes MC, Ledón N, Mazorra Z, Crombet T, Lage A, Bencomo-Hernandez A, Saavedra D. Assessment of non-classical lymphocyte populations in patients with advanced lung cancer treated with Biomodulina T following platinum-based chemotherapy. EXPLORATION OF IMMUNOLOGY 2024; 4:433-445. [DOI: 10.37349/ei.2024.00150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 05/30/2024] [Indexed: 01/03/2025]
Abstract
Aim: Currently, malignant diseases represent a health issue worldwide. Among these, lung cancer is of growing importance, due to its high incidence and mortality. Chemotherapy, one of the most frequently used treatments, has shown its ability to induce accelerated immunosenescence in classic and as well non-classic lymphocyte compartments, being less described in the latter. The immune restoration strategies have demonstrated their ability to reverse immunosenescence and exhaustion markers in conventional lymphocyte subpopulations after chemotherapy. However, the possible immunorestorative effect on non-classical lymphocytes has not been widely reported. The aim of this study was to evaluate the effect of chemotherapy and the administration of a thymic polypeptide factor on non-classical lymphocyte populations in patients with advanced lung cancer.
Methods: Eighteen patients with advanced lung cancer, were evaluated at baseline before and after platinum-based chemotherapy (4–6 cycles). All patients could complete treatment with a thymic polypeptide factor [Biomodulina T (BT)] at the end of chemotherapy. Blood from patients was collected by venipuncture in heparinized tubes before and after chemotherapy and at the end of BT treatment to analyze the frequencies of non-classical immune subpopulations by flow cytometry.
Results: Natural killer (NK), natural killer T cells (NKT), and double-positive T lymphocyte (DPT) proportions reached normal values in patients diagnosed with advanced lung cancer before receiving cytotoxic treatment. Chemotherapy did not induce modifications in the total percent of NK, NKT, and DPT populations in these patients. However, the administration of BT decreased DPTs and NK cells expressing the cluster of differentiation (CD)57 molecule, which is considered a marker of immunosenescence.
Conclusions: These results suggest a lower influence of platinum-based chemotherapy on non-classical lymphocytes and the potential to generate a reconstitution of lymphocyte subpopulations in patients with advanced lung cancer by using the thymic factor BT, which reveals a new possibility for improving the response to cancer immunotherapies [Cuban Public Registry of Clinical Trial (RPCEC, https://rpcec.sld.cu/en/trials/RPCEC00000358-En) identifier: RPCEC00000358].
Collapse
Affiliation(s)
- Gisela María Suárez
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba; Laboratory of Immunology, Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Mauricio Catalá
- Oncology Unit, Medical & Surgical Research Center (CIMEQ), Havana 11300, Cuba
| | - Yadira Peña
- Oncology Unit, Medical & Surgical Research Center (CIMEQ), Havana 11300, Cuba
| | - Susana Portela
- Oncology Unit, Medical & Surgical Research Center (CIMEQ), Havana 11300, Cuba
| | - Ana Laura Añé-Kourí
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| | - Amnely González
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| | | | - Manuel Díaz
- Benéfico-Jurídico Pneumological Hospital, Havana 10600, Cuba
| | | | - Karla Pereira
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| | | | - Mary Carmen Reyes
- Clinical Direction, National Center for Biopreparations, Bejucal, Mayabeque 32600, Cuba
| | - Nuris Ledón
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| | - Zaima Mazorra
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba; Laboratory of Immunology, Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Tania Crombet
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| | - Agustin Lage
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| | | | - Danay Saavedra
- Clinical Immunology Department, Center of Molecular Immunology, Havana 11600, Cuba
| |
Collapse
|
16
|
Cheng KW, Yen CH, Chang R, Wei JCC, Wang SI. Real-World Assessment of Recommended COVID-19 Vaccination Waiting Period after Chemotherapy. Vaccines (Basel) 2024; 12:678. [PMID: 38932407 PMCID: PMC11209144 DOI: 10.3390/vaccines12060678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
There is a knowledge gap concerning the proper timing for COVID-19 vaccination in cancer patients undergoing chemotherapy. We aimed to evaluate the suitability of the guidelines that recommend waiting at least three months after undergoing chemotherapy before receiving a COVID-19 vaccine. This retrospective cohort study used aggregated data from the TriNetX US Collaboratory network. Participants were grouped into two groups based on the interval between chemotherapy and vaccination. The primary outcome assessed was infection risks, including COVID-19; skin, intra-abdominal, and urinary tract infections; pneumonia; and sepsis. Secondary measures included healthcare utilization and all causes of mortality. Kaplan-Meier analysis and the Cox proportional hazard model were used to calculate the cumulative incidence and hazard ratio (HR) and 95% confidence intervals for the outcomes. The proportional hazard assumption was tested with the generalized Schoenfeld approach. Four subgroup analyses (cancer type, vaccine brand, sex, age) were conducted. Sensitivity analyses were performed to account for competing risks and explore three distinct time intervals. Patients receiving a vaccine within three months after chemotherapy had a higher risk of COVID-19 infection (HR: 1.428, 95% CI: 1.035-1.970), urinary tract infection (HR: 1.477, 95% CI: 1.083-2.014), and sepsis (HR: 1.854, 95% CI: 1.091-3.152) compared to those who adhered to the recommendations. Hospital inpatient service utilization risk was also significantly elevated for the within three months group (HR: 1.692, 95% CI: 1.354-2.115). Adhering to a three-month post-chemotherapy waiting period reduces infection and healthcare utilization risks for cancer patients receiving a COVID-19 vaccine.
Collapse
Affiliation(s)
- Kai-Wen Cheng
- Department of Emergency Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Chi-Hua Yen
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Renin Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
- Department of Recreation and Sports Management, Tajen University, Pintung 90741, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Nursing, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40447, Taiwan
- Office of Research and Development, Asia University, Taichung 41354, Taiwan
| | - Shiow-Ing Wang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Center for Health Data Science, Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
17
|
Kientega T, Marcoux S, Bourbonnais J, Montpetit J, Caru M, Cardin GB, Arbour N, Marcil V, Curnier D, Laverdière C, Sinnett D, Rodier F. Premature thymic functional senescence is a hallmark of childhood acute lymphoblastic leukemia survivorship. Blood Cancer J 2024; 14:96. [PMID: 38871704 PMCID: PMC11176394 DOI: 10.1038/s41408-024-01071-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024] Open
Abstract
Childhood acute lymphoblastic leukemia (cALL) survivors suffer early-onset chronic diseases classically associated with aging. Normal aging is accompanied by organ dysfunctions, including immunological ones. We hypothesize that thymic immunosenescence occurs in cALL survivors and that its severity may correlate with early-onset chronic diseases. The PETALE study is a cALL survivor cohort with an extensive cardiovascular and metabolic evaluation. The thymic immunosenescence biomarker, signal joint T-cell receptor excision circles (TREC), was evaluated and was highly correlated with age in healthy participants (n = 281) and cALL survivors (n = 248). We observed a systematic thymic immunoage accentuation in each cALL survivor compared to controls ranging from 5.9 to 88.3 years. The immunoage gain was independent of age at diagnosis and treatment modalities and was more severe for females. Thymic aging was associated with several pathophysiological parameters, was greater in survivors suffering from metabolic syndrome, but there was no significant association with global physical condition. The decrease in TREC was independent from blood cell counts, which were normal, suggesting a segmental aging of the thymic compartment. Indeed, increased plasmatic T cell regulatory cytokines IL-6, IL-7 and GM-CSF accompanied high immunoage gain. Our data reveal that cALL or its treatment trigger a rapid immunoage gain followed by further gradual thymic immunosenescence, similar to normal aging. This leads to an enduring shift in accentuated immunoage compared to chronological age. Thus, accentuated thymic immunosenescence is a hallmark of cALL survivorship and TREC levels could be useful immunosenescence biomarkers to help monitoring the health of cancer survivors.
Collapse
Affiliation(s)
- Tibila Kientega
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Sophie Marcoux
- Université Laval, Département de médecine sociale et préventive, Québec, QC, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Jessica Bourbonnais
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Jade Montpetit
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Maxime Caru
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Department of Pediatrics, Division of Hematology and Oncology, Penn State College of Medicine, Hershey, PA, USA
| | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Nathalie Arbour
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Université de Montréal, Département de Neurosciences, Montréal, QC, Canada
| | - Valérie Marcil
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Département de Nutrition, Montréal, QC, Canada
| | - Daniel Curnier
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Faculté de médecine, École de kinésiologie et des sciences de l'activité physique, Laboratoire de physiopathologie de l'exercice (LPEX), Montréal, QC, Canada
| | - Caroline Laverdière
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Département de Pédiatrie, Montréal, QC, Canada
| | - Daniel Sinnett
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Département de Pédiatrie, Montréal, QC, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada.
- Institut du cancer de Montréal, Montréal, QC, Canada.
- Université de Montréal, Département de Radiologie, radio-oncologie et médecine nucléaire, Montréal, QC, Canada.
| |
Collapse
|
18
|
Gkekos L, Johansson ALV, Rodriguez-Wallberg KA, Fredriksson I, Lundberg FE. Obstetric and perinatal outcomes in women with previous breast cancer: a nationwide study of singleton births 1973-2017. Hum Reprod Open 2024; 2024:hoae027. [PMID: 38784055 PMCID: PMC11112047 DOI: 10.1093/hropen/hoae027] [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: 10/05/2023] [Revised: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
STUDY QUESTION What are the obstetric and perinatal outcomes in births to breast cancer survivors compared to women without previous breast cancer? SUMMARY ANSWER Women who conceived during the first 2 years following a breast cancer diagnosis had a higher risk for preterm birth, induced delivery, and cesarean section, while no increased risks were observed in births conceived later than 2 years after a breast cancer diagnosis. WHAT IS KNOWN ALREADY A recent meta-analysis found higher risks of cesarean section, preterm birth, low birthweight, and small for gestational age in pregnancies among breast cancer survivors. Less is known about rarer outcomes such as pre-eclampsia or congenital malformations. STUDY DESIGN SIZE DURATION We conducted a population-based matched cohort study including all breast cancer survivors who gave birth to singletons 1973-2017 in Sweden, identified through linkage between the Swedish Cancer Register, the Medical Birth Register, and the National Quality Register for Breast Cancer. PARTICIPANTS/MATERIALS SETTINGS METHODS Each birth following breast cancer (n = 926) was matched by maternal age at delivery, parity, and calendar year at delivery to 100 births in a comparator cohort of women (n = 92 490). Conditional logistic and multinomial regression models estimated relative risks (RR) with 95% CI. Subgroup analyses by time since diagnosis and type of treatment were performed. MAIN RESULTS AND THE ROLE OF CHANCE Previous breast cancer was associated with higher risks of induced delivery (RR; 1.3, 1.0-1.6), very preterm birth (RR; 1.8, 1.1-3.0), and planned preterm birth (RR; 1.6, 1.0-2.4). Women who conceived within 1 year after breast cancer diagnosis had higher risks of cesarean section (RR; 1.7, 1.0-2.7), very preterm birth (RR; 5.3, 1.9-14.8), and low birthweight (RR; 2.7, 1.4-5.2), while the risks of induced delivery (RR; 1.8, 1.1-2.9), moderately preterm birth (RR; 2.1, 1.2-3.7), and planned preterm birth (RR; 2.5, 1.1-5.7) were higher in women who conceived during the second year after diagnosis. Women who conceived later than 2 years after breast cancer diagnosis had similar obstetric risks to their comparators. LIMITATIONS REASONS FOR CAUTION As information on the end date of treatment was unavailable, the time between the date of diagnosis and conception was used as a proxy, which does not fully capture the effect of time since end of treatment. In addition, treatments and clinical recommendations have changed over the long study period, which may impact childbearing patterns in breast cancer survivors. WIDER IMPLICATIONS OF THE FINDINGS Risks of adverse obstetric outcomes in breast cancer survivors were confined to births conceived within 2 years of diagnosis. As family building holds significance for numerous young breast cancer patients, these findings are particularly important to inform both breast cancer survivors and clinicians about future reproductive outcomes. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Swedish Cancer Society (grant number 22-2044 Pj A.L.V.J.), Karolinska Institutet Foundations (grant number: 2022-01696 F.E.L., 2022-01559 A.L.V.J.), and the Swedish Research Council (grant number: 2021-01657 A.L.V.J.). K.A.R.-W. is supported by grants from the Swedish Cancer Society (20 0170 F) and the Radiumhemmets Research Foundations for clinical researchers 2020-2026. The authors declare that they have no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Leo Gkekos
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anna L V Johansson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Laboratory of Translational Fertility Preservation, Karolinska Institutet, Stockholm, Sweden
- Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Irma Fredriksson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Frida E Lundberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology-Pathology, Laboratory of Translational Fertility Preservation, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Dai L, Tan Q, Li L, Lou N, Zheng C, Yang J, Huang L, Wang S, Luo R, Fan G, Xie T, Yao J, Zhang Z, Tang L, Shi Y, Han X. High-Throughput Antigen Microarray Identifies Longitudinal Prognostic Autoantibody for Chemoimmunotherapy in Advanced Non-Small Cell Lung Cancer. Mol Cell Proteomics 2024; 23:100749. [PMID: 38513890 PMCID: PMC11070596 DOI: 10.1016/j.mcpro.2024.100749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/03/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Chemoimmunotherapy has evolved as a standard treatment for advanced non-small cell lung cancer (aNSCLC). However, inevitable drug resistance has limited its efficacy, highlighting the urgent need for biomarkers of chemoimmunotherapy. A three-phase strategy to discover, verify, and validate longitudinal predictive autoantibodies (AAbs) for aNSCLC before and after chemoimmunotherapy was employed. A total of 528 plasma samples from 267 aNSCLC patients before and after anti-PD1 immunotherapy were collected, plus 30 independent formalin-fixed paraffin-embedded samples. Candidate AAbs were firstly selected using a HuProt high-density microarray containing 21,000 proteins in the discovery phase, followed by validation using an aNSCLC-focused microarray. Longitudinal predictive AAbs were chosen for ELISA based on responders versus non-responders comparison and progression-free survival (PFS) survival analysis. Prognostic markers were also validated using immunohistochemistry and publicly available immunotherapy datasets. We identified and validated a panel of two AAbs (MAX and DHX29) as pre-treatment biomarkers and another panel of two AAbs (MAX and TAPBP) as on-treatment predictive markers in aNSCLC patients undergoing chemoimmunotherapy. All three AAbs exhibited a positive correlation with early responses and PFS (p < 0.05). The kinetics of MAX AAb showed an increasing trend in responders (p < 0.05) and a tendency to initially increase and then decrease in non-responders (p < 0.05). Importantly, MAX protein and mRNA levels effectively discriminated PFS (p < 0.05) in aNSCLC patients treated with immunotherapy. Our results present a longitudinal analysis of changes in prognostic AAbs in aNSCLC patients undergoing chemoimmunotherapy.
Collapse
Affiliation(s)
- Liyuan Dai
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Qiaoyun Tan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ning Lou
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Cuiling Zheng
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jianliang Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Liling Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Shasha Wang
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Rongrong Luo
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Guangyu Fan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Zhishang Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
20
|
Chauhan SK, Dunn C, Andresen NK, Røssevold AH, Skorstad G, Sike A, Gilje B, Raj SX, Huse K, Naume B, Kyte JA. Peripheral immune cells in metastatic breast cancer patients display a systemic immunosuppressed signature consistent with chronic inflammation. NPJ Breast Cancer 2024; 10:30. [PMID: 38653982 DOI: 10.1038/s41523-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Immunotherapies blocking the PD-1/PD-L1 checkpoint show some efficacy in metastatic breast cancer (mBC) but are often hindered by immunosuppressive mechanisms. Understanding these mechanisms is crucial for personalized treatments, with peripheral blood monitoring representing a practical alternative to repeated biopsies. In the present study, we performed a comprehensive mass cytometry analysis of peripheral blood immune cells in 104 patients with HER2 negative mBC and 20 healthy donors (HD). We found that mBC patients had significantly elevated monocyte levels and reduced levels of CD4+ T cells and plasmacytoid dendritic cells, when compared to HD. Furthermore, mBC patients had more effector T cells and regulatory T cells, increased expression of immune checkpoints and other activation/exhaustion markers, and a shift to a Th2/Th17 phenotype. Furthermore, T-cell phenotypes identified by mass cytometry correlated with functionality as assessed by IFN-γ production. Additional analysis indicated that previous chemotherapy and CDK4/6 inhibition impacted the numbers and phenotype of immune cells. From 63 of the patients, fresh tumor samples were analyzed by flow cytometry. Paired PBMC-tumor analysis showed moderate correlations between peripheral CD4+ T and NK cells with their counterparts in tumors. Further, a CD4+ T cell cluster in PBMCs, that co-expressed multiple checkpoint receptors, was negatively associated with CD4+ T cell tumor infiltration. In conclusion, the identified systemic immune signatures indicate an immune-suppressed environment in mBC patients who had progressed/relapsed on standard treatments, and is consistent with ongoing chronic inflammation. These activated immuno-suppressive mechanisms may be investigated as therapeutic targets, and for use as biomarkers of response or treatment resistance.
Collapse
Affiliation(s)
- Sudhir Kumar Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Claire Dunn
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Nikolai Kragøe Andresen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gjertrud Skorstad
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Adam Sike
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Sunil Xavier Raj
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
21
|
Sevieri M, Andreata F, Mainini F, Signati L, Piccotti F, Truffi M, Bonizzi A, Sitia L, Pigliacelli C, Morasso C, Tagliaferri B, Corsi F, Mazzucchelli S. Impact of doxorubicin-loaded ferritin nanocages (FerOX) vs. free doxorubicin on T lymphocytes: a translational clinical study on breast cancer patients undergoing neoadjuvant chemotherapy. J Nanobiotechnology 2024; 22:184. [PMID: 38622644 PMCID: PMC11020177 DOI: 10.1186/s12951-024-02441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Despite the advent of numerous targeted therapies in clinical practice, anthracyclines, including doxorubicin (DOX), continue to play a pivotal role in breast cancer (BC) treatment. DOX directly disrupts DNA replication, demonstrating remarkable efficacy against BC cells. However, its non-specificity toward cancer cells leads to significant side effects, limiting its clinical utility. Interestingly, DOX can also enhance the antitumor immune response by promoting immunogenic cell death in BC cells, thereby facilitating the presentation of tumor antigens to the adaptive immune system. However, the generation of an adaptive immune response involves highly proliferative processes, which may be adversely affected by DOX-induced cytotoxicity. Therefore, understanding the impact of DOX on dividing T cells becomes crucial, to deepen our understanding and potentially devise strategies to shield anti-tumor immunity from DOX-induced toxicity. Our investigation focused on studying DOX uptake and its effects on human lymphocytes. We collected lymphocytes from healthy donors and BC patients undergoing neoadjuvant chemotherapy (NAC). Notably, patient-derived peripheral blood mononuclear cells (PBMC) promptly internalized DOX when incubated in vitro or isolated immediately after NAC. These DOX-treated PBMCs exhibited significant proliferative impairment compared to untreated cells or those isolated before treatment initiation. Intriguingly, among diverse lymphocyte sub-populations, CD8 + T cells exhibited the highest uptake of DOX. To address this concern, we explored a novel DOX formulation encapsulated in ferritin nanocages (FerOX). FerOX specifically targets tumors and effectively eradicates BC both in vitro and in vivo. Remarkably, only T cells treated with FerOX exhibited reduced DOX internalization, potentially minimizing cytotoxic effects on adaptive immunity.Our findings underscore the importance of optimizing DOX delivery to enhance its antitumor efficacy while minimizing adverse effects, highlighting the pivotal role played by FerOX in mitigating DOX-induced toxicity towards T-cells, thereby positioning it as a promising DOX formulation. This study contributes valuable insights to modern cancer therapy and immunomodulation.
Collapse
Affiliation(s)
- Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
| | - Francesco Andreata
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Mainini
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | | | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - Arianna Bonizzi
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy
| | - Claudia Pigliacelli
- Laboratory of Supramolecular and Bio-Nanomaterials (SBNLab), Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, 20131, Italy
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | | | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy.
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy.
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan, 20157, Italy.
| |
Collapse
|
22
|
Turpin R, Liu R, Munne PM, Peura A, Rannikko JH, Philips G, Boeckx B, Salmelin N, Hurskainen E, Suleymanova I, Aung J, Vuorinen EM, Lehtinen L, Mutka M, Kovanen PE, Niinikoski L, Meretoja TJ, Mattson J, Mustjoki S, Saavalainen P, Goga A, Lambrechts D, Pouwels J, Hollmén M, Klefström J. Respiratory complex I regulates dendritic cell maturation in explant model of human tumor immune microenvironment. J Immunother Cancer 2024; 12:e008053. [PMID: 38604809 PMCID: PMC11015234 DOI: 10.1136/jitc-2023-008053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Combining cytotoxic chemotherapy or novel anticancer drugs with T-cell modulators holds great promise in treating advanced cancers. However, the response varies depending on the tumor immune microenvironment (TIME). Therefore, there is a clear need for pharmacologically tractable models of the TIME to dissect its influence on mono- and combination treatment response at the individual level. METHODS Here we establish a patient-derived explant culture (PDEC) model of breast cancer, which retains the immune contexture of the primary tumor, recapitulating cytokine profiles and CD8+T cell cytotoxic activity. RESULTS We explored the immunomodulatory action of a synthetic lethal BCL2 inhibitor venetoclax+metformin drug combination ex vivo, discovering metformin cannot overcome the lymphocyte-depleting action of venetoclax. Instead, metformin promotes dendritic cell maturation through inhibition of mitochondrial complex I, increasing their capacity to co-stimulate CD4+T cells and thus facilitating antitumor immunity. CONCLUSIONS Our results establish PDECs as a feasible model to identify immunomodulatory functions of anticancer drugs in the context of patient-specific TIME.
Collapse
Affiliation(s)
- Rita Turpin
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Ruixian Liu
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Pauliina M Munne
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Aino Peura
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | | | | | - Bram Boeckx
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Natasha Salmelin
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Elina Hurskainen
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Ilida Suleymanova
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - July Aung
- University of Helsinki Faculty of Medicine, Helsinki, Finland
| | | | | | - Minna Mutka
- Department of Pathology, Helsinki University Central Hospital, Helsinki, Finland
| | - Panu E Kovanen
- Department of Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Laura Niinikoski
- Breast Surgery Unit, Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Tuomo J Meretoja
- Breast Surgery Unit, Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Johanna Mattson
- Department of oncology, Helsinki University Central Hospital, Helsinki, Finland
| | - Satu Mustjoki
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- University of Helsinki Helsinki Institute of Life Sciences, Helsinki, Finland
| | | | - Andrei Goga
- Department of Cell & Tissue Biology, UCSF, San Francisco, California, USA
| | | | - Jeroen Pouwels
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | | | - Juha Klefström
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
- Finnish Cancer Institute, Helsinki, Finland
| |
Collapse
|
23
|
Rupp L, Dietsche I, Kießler M, Sommer U, Muckenhuber A, Steiger K, van Eijck CWF, Richter L, Istvanffy R, Jäger C, Friess H, van Eijck CHJ, Demir IE, Reyes CM, Schmitz M. Neoadjuvant chemotherapy is associated with suppression of the B cell-centered immune landscape in pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1378190. [PMID: 38629072 PMCID: PMC11018975 DOI: 10.3389/fimmu.2024.1378190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at advanced stages and associated with early distant metastasis and poor survival. Besides clinical factors, the tumor microenvironment (TME) emerged as a crucial determinant of patient survival and therapy response in many tumors, including PDAC. Thus, the presence of tumor-infiltrating lymphocytes and the formation of tertiary lymphoid structures (TLS) is associated with longer survival in PDAC. Although neoadjuvant therapy (NeoTx) has improved the management of locally advanced tumors, detailed insight into its effect on various TME components is limited. While a remodeling towards a proinflammatory state was reported for PDAC-infiltrating T cells, the effect of NeoTx on B cell subsets, including plasma cells, and TLS formation is widely unclear. We thus investigated the frequency, composition, and spatial distribution of PDAC-infiltrating B cells in primary resected (PR) versus neoadjuvant-treated patients using a novel multiplex immunohistochemistry panel. The NeoTx group displayed significantly lower frequencies of pan B cells, GC B cells, plasmablasts, and plasma cells, accompanied by a reduced abundance of TLS. This finding was supported by bulk RNA-sequencing analysis of an independent fresh frozen tissue cohort, which revealed that major B cell pathways were downregulated in the NeoTx group. We further observed that plasma cells frequently formed aggregates that localized close to TLS and that TLS+ patients displayed significantly higher plasma cell frequencies compared to TLS- patients in the PR group. Additionally, high densities of CD20+ intratumoral B cells were significantly associated with longer overall survival in the PR group. While CD20+ B cells held no prognostic value for NeoTx patients, an increased frequency of proliferating CD20+Ki67+ B cells emerged as an independent prognostic factor for longer survival in the NeoTx group. These results indicate that NeoTx differentially affects PDAC-infiltrating immune cells and may have detrimental effects on the existing B cell landscape and the formation of TLS. Gaining further insight into the underlying molecular mechanisms is crucial to overcome the intrinsic immunotherapy resistance of PDAC and develop novel strategies to improve the long-term outcome of PDAC patients.
Collapse
Affiliation(s)
- Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ina Dietsche
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Maximilian Kießler
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Alexander Muckenhuber
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Casper W. F. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Leonard Richter
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Casper H. J. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of General Surgery, Hepato-Pancreato-Biliary (HPB) Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
- Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, Technical University of Munich, Munich, Germany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
24
|
Bar J, Leibowitz R, Reinmuth N, Ammendola A, Jacob E, Moskovitz M, Levy-Barda A, Lotem M, Katsenelson R, Agbarya A, Abu-Amna M, Gottfried M, Harkovsky T, Wolf I, Tepper E, Loewenthal G, Yellin B, Brody Y, Dahan N, Yanko M, Lahav C, Harel M, Raveh Shoval S, Elon Y, Sela I, Dicker AP, Shaked Y. Biological insights from plasma proteomics of non-small cell lung cancer patients treated with immunotherapy. Front Immunol 2024; 15:1364473. [PMID: 38487531 PMCID: PMC10937428 DOI: 10.3389/fimmu.2024.1364473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/02/2024] [Indexed: 03/17/2024] Open
Abstract
Introduction Immune checkpoint inhibitors have made a paradigm shift in the treatment of non-small cell lung cancer (NSCLC). However, clinical response varies widely and robust predictive biomarkers for patient stratification are lacking. Here, we characterize early on-treatment proteomic changes in blood plasma to gain a better understanding of treatment response and resistance. Methods Pre-treatment (T0) and on-treatment (T1) plasma samples were collected from 225 NSCLC patients receiving PD-1/PD-L1 inhibitor-based regimens. Plasma was profiled using aptamer-based technology to quantify approximately 7000 plasma proteins per sample. Proteins displaying significant fold changes (T1:T0) were analyzed further to identify associations with clinical outcomes using clinical benefit and overall survival as endpoints. Bioinformatic analyses of upregulated proteins were performed to determine potential cell origins and enriched biological processes. Results The levels of 142 proteins were significantly increased in the plasma of NSCLC patients following ICI-based treatments. Soluble PD-1 exhibited the highest increase, with a positive correlation to tumor PD-L1 status, and, in the ICI monotherapy dataset, an association with improved overall survival. Bioinformatic analysis of the ICI monotherapy dataset revealed a set of 30 upregulated proteins that formed a single, highly interconnected network, including CD8A connected to ten other proteins, suggestive of T cell activation during ICI treatment. Notably, the T cell-related network was detected regardless of clinical benefit. Lastly, circulating proteins of alveolar origin were identified as potential biomarkers of limited clinical benefit, possibly due to a link with cellular stress and lung damage. Conclusions Our study provides insights into the biological processes activated during ICI-based therapy, highlighting the potential of plasma proteomics to identify mechanisms of therapy resistance and biomarkers for outcome.
Collapse
Affiliation(s)
- Jair Bar
- Institute of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Raya Leibowitz
- Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Shamir Medical Center, Oncology Institute, Zerifin, Israel
| | - Niels Reinmuth
- German Center for Lung Research (DZL), Munich-Gauting, Germany
- Biobank of lung disease, Asklepios Klinik Gauting GmbH, Gauting, Germany
| | - Astrid Ammendola
- Biobank of lung disease, Asklepios Klinik Gauting GmbH, Gauting, Germany
| | | | - Mor Moskovitz
- Thoracic oncology service, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Adva Levy-Barda
- Biobank, Department of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Michal Lotem
- Center for Melanoma and Cancer Immunotherapy, Hadassah Hebrew University Medical Center, Sharett Institute of Oncology, Jerusalem, Israel
| | | | - Abed Agbarya
- Institute of Oncology, Bnai Zion Medical Center, Haifa, Israel
| | - Mahmoud Abu-Amna
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, Afula, Israel
| | - Maya Gottfried
- Department of Oncology, Meir Medical Center, Kfar-Saba, Israel
| | - Tatiana Harkovsky
- Barzilai Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Ashkelon, Israel
| | - Ido Wolf
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ella Tepper
- Department of Oncology, Assuta Hospital, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | | - Adam P. Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Yuval Shaked
- Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
25
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Naumenko VA, Chekhonin VP. The need for paradigm shift: prognostic significance and implications of standard therapy-related systemic immunosuppression in glioblastoma for immunotherapy and oncolytic virotherapy. Front Immunol 2024; 15:1326757. [PMID: 38390330 PMCID: PMC10881776 DOI: 10.3389/fimmu.2024.1326757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Victor A. Naumenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
26
|
Kresovich JK, O’Brien KM, Xu Z, Weinberg CR, Sandler DP, Taylor JA. Circulating Leukocyte Subsets Before and After a Breast Cancer Diagnosis and Therapy. JAMA Netw Open 2024; 7:e2356113. [PMID: 38358741 PMCID: PMC10870180 DOI: 10.1001/jamanetworkopen.2023.56113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Importance Changes in leukocyte composition often precede chronic disease onset. Patients with a history of breast cancer (hereinafter referred to as breast cancer survivors) are at increased risk for subsequent chronic diseases, but the long-term changes in peripheral leukocyte composition following a breast cancer diagnosis and treatment remain unknown. Objective To examine longitudinal changes in peripheral leukocyte composition in women who did and did not develop breast cancer and identify whether differences in breast cancer survivors were associated with specific treatments. Design, Setting, and Participants In this prospective cohort study, paired blood samples were collected from 2315 women enrolled in The Sister Study, a US-nationwide prospective cohort study of 50 884 women, at baseline (July 2003 to March 2009) and follow-up (October 2013 to March 2015) home visits, with a mean (SD) follow-up interval of 7.6 (1.4) years. By design, approximately half of the included women had been diagnosed and treated for breast cancer after enrollment and before the second blood draw. A total of 410 women were included in the present study, including 185 breast cancer survivors and 225 who remained free of breast cancer over a comparable follow-up period. Data were analyzed from April 21 to September 9, 2022. Exposures Breast cancer status and, among breast cancer survivors, cancer treatment type (chemotherapy, radiotherapy, endocrine therapy, or surgery). Main Outcomes and Measures Blood DNA methylation data were generated in 2019 using a genome-wide methylation screening tool and deconvolved to estimate percentages of 12 circulating leukocyte subsets. Results Of the 410 women included in the analysis, the mean (SD) age at enrollment was 56 (9) years. Compared with breast cancer-free women, breast cancer survivors had decreased percentages of circulating eosinophils (-0.45% [95% CI, -0.87% to -0.03%]; P = .03), total CD4+ helper T cells (-1.50% [95% CI, -2.56% to -0.44%]; P = .01), and memory B cells (-0.22% [95% CI, -0.34% to -0.09%]; P = .001) and increased percentages of circulating naive B cells (0.46% [95% CI, 0.17%-0.75%]; P = .002). In breast cancer survivor-only analyses, radiotherapy was associated with decreases in total CD4+ T cell levels, whereas chemotherapy was associated with increases in naive B cell levels. Surgery and endocrine therapy were not meaningfully associated with leukocyte changes. Conclusions and Relevance In this cohort study of 410 women, breast cancer survivors experienced lasting changes in peripheral leukocyte composition compared with women who remained free of breast cancer. These changes may be related to treatment with chemotherapy or radiotherapy and could influence future chronic disease risk.
Collapse
Affiliation(s)
- Jacob K. Kresovich
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Breast Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Research Triangle Park, North Carolina
| | - Katie M. O’Brien
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Research Triangle Park, North Carolina
| | - Zongli Xu
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Research Triangle Park, North Carolina
| | - Clarice R. Weinberg
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| | - Dale P. Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Research Triangle Park, North Carolina
| | - Jack A. Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Research Triangle Park, North Carolina
- Epigenetic and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| |
Collapse
|
27
|
Eterafi M, Fouladi N, Golizadeh M, Shaker H, Matin S, Safarzadeh E. Reported side-effects following Oxford/AstraZeneca COVID-19 vaccine in the north-west province, Iran: A cross-sectional study. PLoS One 2024; 19:e0296669. [PMID: 38181026 PMCID: PMC10769020 DOI: 10.1371/journal.pone.0296669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/17/2023] [Indexed: 01/07/2024] Open
Abstract
While the vaccination was introduced as a promising tool to control the Coronavirus disease 2019 (COVID-19) pandemic, concerns about vaccine-related side effects had grown. Due to the widespread administration of the COVID-19 vaccine worldwide for the first time, it was necessary to evaluate the safety and potential side effects in recipients. This study aims to assess, the incidence of adverse effects following Oxford-AstraZeneca vaccination and identify their related factors. In this cross-sectional survey-based study, 453 volunteers participated, including 235 men and 218 women. The reported adverse reactions from recipients of the ChAdOx1 nCoV-19 (Oxford-AstraZeneca) vaccine were collected by using a questionnaire. The findings showed that the incidence of adverse reactions, such as neurological, systematic, gastrointestinal, respiratory, and local symptoms were significantly higher after the first dose compared to the second dose. Systematic symptoms were the most prevalent reported side effects after the first and second dose injection. The demographical study of participants showed that individuals aged 18-34 and females were more prone to present adverse events following vaccination. However, no significant relationship was found between the occurrence of side effects and the recipients' body mass index. Despite the life-saving role of vaccination against SARS-CoV-2, it may have some adverse reactions in recipients. The severity and frequency of side effects were different. So, they were dependent on several factors, including gender and age. Altogether, post-vaccination adverse reactions were mild and tolerable.
Collapse
Affiliation(s)
- Majid Eterafi
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nasrin Fouladi
- School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
- Social Determinants of Health Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Golizadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamidreza Shaker
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Somaieh Matin
- Department of Internal Medicine, Emam Khomeini Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
- Gastrointestinal and Liver Disease Research Center, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
28
|
Heiser RA, Cao AT, Zeng W, Ulrich M, Younan P, Anderson ME, Trueblood ES, Jonas M, Thurman R, Law CL, Gardai SJ. Brentuximab Vedotin-Driven Microtubule Disruption Results in Endoplasmic Reticulum Stress Leading to Immunogenic Cell Death and Antitumor Immunity. Mol Cancer Ther 2024; 23:68-83. [PMID: 37775098 PMCID: PMC10762337 DOI: 10.1158/1535-7163.mct-23-0118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/07/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Brentuximab vedotin, a CD30-directed antibody-drug conjugate (ADC), is approved for clinical use in multiple CD30-expressing lymphomas. The cytotoxic payload component of brentuximab vedotin is monomethyl auristatin E (MMAE), a highly potent microtubule-disrupting agent. Preclinical results provided here demonstrate that treatment of cancer cells with brentuximab vedotin or free MMAE leads to a catastrophic disruption of the microtubule network eliciting a robust endoplasmic reticulum (ER) stress response that culminates in the induction of the classic hallmarks of immunogenic cell death (ICD). In accordance with the induction of ICD, brentuximab vedotin-killed lymphoma cells drove innate immune cell activation in vitro and in vivo. In the "gold-standard" test of ICD, vaccination of mice with brentuximab vedotin or free MMAE-killed tumor cells protected animals from tumor rechallenge; in addition, T cells transferred from previously vaccinated animals slowed tumor growth in immunodeficient mice. Immunity acquired from killed tumor cell vaccination was further amplified by the addition of PD-1 blockade. In a humanized model of CD30+ B-cell tumors, treatment with brentuximab vedotin drove the expansion and recruitment of autologous Epstein-Barr virus-reactive CD8+ T cells potentiating the activity of anti-PD-1 therapy. Together, these data support the ability of brentuximab vedotin and MMAE to drive ICD in tumor cells resulting in the activation of antigen-presenting cells and augmented T-cell immunity. These data provide a strong rationale for the clinical combination of brentuximab vedotin and other MMAE-based ADCs with checkpoint inhibitors.
Collapse
|
29
|
Wimmer K, Sachet M, Ramos C, Frantal S, Birnleitner H, Brostjan C, Exner R, Filipits M, Bago-Horvath Z, Rudas M, Bartsch R, Gnant M, Singer CF, Balic M, Egle D, Oehler R, Fitzal F. Differential immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel in breast cancer patients. J Exp Clin Cancer Res 2023; 42:300. [PMID: 37957750 PMCID: PMC10644559 DOI: 10.1186/s13046-023-02876-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Epirubicin/cyclophosphamide (EC) and docetaxel (D) are commonly used in a sequential regimen in the neoadjuvant treatment of early, high-risk or locally advanced breast cancer (BC). Novel approaches to increase the response rate combine this treatment with immunotherapies such as PD-1 inhibition. However, the expected stimulatory effect on lymphocytes may depend on the chemotherapy backbone. Therefore, we separately compared the immunomodulatory effects of EC and D in the setting of a randomized clinical trial. METHODS Tumor and blood samples of 154 patients from the ABCSG-34 trial were available (76 patients received four cycles of EC followed by four cycles of D; 78 patients get the reverse treatment sequence). Tumor-infiltrating lymphocytes, circulating lymphocytes and 14 soluble immune mediators were determined at baseline and at drug change. Furthermore, six BC cell lines were treated with E, C or D and co-cultured with immune cells. RESULTS Initial treatment with four cycles of EC reduced circulating B and T cells by 94% and 45%, respectively. In contrast, no comparable effects on lymphocytes were observed in patients treated with initial four cycles of D. Most immune mediators decreased under EC whereas D-treatment resulted in elevated levels of CXCL10, urokinase-type plasminogen activator (uPA) and its soluble receptor (suPAR). Accordingly, only the exposure of BC cell lines to D induced similar increases as compared to E. While treatment of BC cells with E was associated with cell shrinkage and apoptosis, D induced cell swelling and accumulation of cells in G2 phase. CONCLUSION The deleterious effect of EC on lymphocytes indicates strong immunosuppressive properties of this combination therapy. D, in contrast, has no effect on lymphocytes, but triggers the secretion of stimulatory proteins in vivo and in vitro, indicating a supportive effect on the immune system. Underlying differences in the induced cell death might be causal. These divergent immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel should be considered when planning future combinations with immunotherapies in breast cancer.
Collapse
Affiliation(s)
- Kerstin Wimmer
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Monika Sachet
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Sophie Frantal
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Hanna Birnleitner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ruth Exner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Martin Filipits
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Margaretha Rudas
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Rupert Bartsch
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Gnant
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Christian F Singer
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
- Department of Gynecology, Medical University of Vienna, 1090, Vienna, Austria
| | - Marija Balic
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Oncology, Medical University of Graz, Graz, Austria
| | - Daniel Egle
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria.
| | - Florian Fitzal
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| |
Collapse
|
30
|
Tseng I, Ai D, Chen Y, Zhu H, Li F, Xu Y, Yu L, Liu Q, Deng J, Hao S, Zhu Z, Zhao W, Fan M, Li L, Su F, Zhao K. Lymphocyte recovery from radiation-induced lymphopenia in locally advanced esophageal squamous cell carcinoma: correlations with prognosis and lymphocyte-related organs. Radiat Oncol 2023; 18:172. [PMID: 37858121 PMCID: PMC10588237 DOI: 10.1186/s13014-023-02354-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Limited studies explored the relationship between lymphocyte recovery after definitive concurrent chemoradiotherapy (dCCRT) and prognosis in esophageal squamous cell carcinoma (ESCC). METHODS ESCC patients with obtainable absolute lymphocyte counts (ALCs) at 6 months after dCCRT were screened from prospective trials. Patients were divided into groups according to the grade of ALC nadir during radiotherapy (G4 or G1-3) and lymphocyte recovery status, which was assessed by lymphocyte recovery index (LRI), calculated as the ratio of post- to pre-treatment lymphocyte counts. Cox analysis was conducted to evaluate the prognostic significance of lymphocyte recovery status. Irradiated relative volumes of the bone marrow (BM) and spleen and effective dose to immune cells (EDIC) were collected to identify their impacts on lymphocyte recovery status by logistic analysis. RESULTS 232 patients were enrolled. In 69 patients with G4 ALC nadir (group A and B) and 163 patients with G1-3 ALC nadir (group C and D) during dCCRT, 27 (group A) and 67 (group C) patients showed an insufficient level of lymphocyte recovery (LRI < 60%), and 42 (group B) and 96 (group D) patients showed a satisfactory level of lymphocyte recovery (LRI ≥ 60%). Cox multivariable analysis revealed that inadequate lymphocyte recovery was significantly associated with worse overall survival (HR, 2.80 and 1.70) and local recurrence-free survival (HR, 2.82 and 1.60) both in group A vs group B and group C vs group D. Logistic analysis identified BM V5 (OR 4.24 and 2.29) as an independent predictor of inadequate lymphocyte recovery from G4 or G1-3 ALC nadir, respectively. CONCLUSIONS Insufficient lymphocyte recovery might serve as a valuable prognostic factor, regardless of whether patients experienced G4 or G1-3 ALC nadir during radiotherapy. Additionally, it was observed that a larger relative volume of BM receiving ≥ 5 Gy was correlated with a higher risk of insufficient lymphocyte recovery.
Collapse
Affiliation(s)
- Ihsuan Tseng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Dashan Ai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Yun Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Hongcheng Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Fangfang Li
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yang Xu
- Department of Medicine, Enhance Human Health Through Pharma Technology Innovation, Shanghai, 201800, China
| | - Lu Yu
- Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Qi Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Jiaying Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Shengnan Hao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Weixin Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Min Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Ling Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Fengtao Su
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| |
Collapse
|
31
|
Schlecht S, Neubert S, Meng K, Rabe A, Jentschke E. Changes of Symptoms of Anxiety, Depression, and Fatigue in Cancer Patients 3 Months after a Video-Based Intervention. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6933. [PMID: 37887671 PMCID: PMC10606592 DOI: 10.3390/ijerph20206933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/23/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023]
Abstract
During the COVID-19 pandemic, social distancing restricted psycho-oncological care. Therefore, this secondary analysis examines the changes in anxiety, fear of progression, fatigue, and depression in cancer patients after a video-based eHealth intervention. We used a prospective observational design with 155 cancer patients with mixed tumor entities. Data were assessed before and after the intervention and at a three-month follow-up using self-reported questionnaires (GAD-7, FOP-Q-SF, PHQ-8, and EORTC QLQ-FA12). The eight videos included psychoeducation, Acceptance and Commitment Therapy elements, and yoga and qigong exercises. The results showed that three months after finishing the video-based intervention, participants showed significantly reduced fear of progression (d = -0.23), depression (d = -0.27), and fatigue (d = -0.24) compared to the baseline. However, there was no change in anxiety (d = -0.09). Findings indicated marginal improvements in mental distress when using video-based intervention for cancer patients for up to three months, but long-term effectiveness must be confirmed using a controlled design.
Collapse
Affiliation(s)
| | | | | | | | - Elisabeth Jentschke
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080 Würzburg, Germany; (S.S.); (S.N.); (K.M.); (A.R.)
| |
Collapse
|
32
|
Li R, Hao Y, Roche K, Chen G, Pan W, Wang AZ, Min Y. Chemotherapy-induced nanovaccines implement immunogenicity equivalence for improving cancer chemoimmunotherapy. Biomaterials 2023; 301:122290. [PMID: 37643488 DOI: 10.1016/j.biomaterials.2023.122290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/28/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
Several chemoimmunotherapies have been approved by the FDA for the treatment of various cancers. Chemotherapy has the potential to improve the efficacy of immunotherapy by inducing immunogenic cell death (ICD) of tumor cells, promoting the release of tumor associated antigens (TAAs), tumor specific antigens (TSAs) and damage associated molecular patterns (DAMPs), and disrupting immunosuppressive microenvironments by tumor debulking. Unfortunately, systemic administration of chemotherapeutics carries side effects of blunting anti-cancer immune response through systemic immunosuppression, which deserves to be explored as an inner contradiction in chemoimmunotherapy. Here, we proposed the hypothesis of "immunogenicity equivalence" in chemoimmunotherapy that chemotherapeutics-induced immunogenic antigens and DAMPs in vitro that can subsequently be incorporated into nanovaccines, which will possess comparable immunostimulatory potential when compared to tumors treated with systemic chemotherapy in vivo. The proteomic analysis confirmed that our nanovaccines contained TAAs, TSAs and DAMPs. Improvement in treatment outcomes in tumor-bearing mice receiving anti-PD-1 and chemotherapy-induced nanovaccines was then observed. Furthermore, we demonstrated the feasibility of replacing long-term chemotherapy with nanovaccines in chemoimmunotherapy. Our nanovaccine strategy would be a general choice for formulating cancer vaccines in personalized medicine.
Collapse
Affiliation(s)
- Rui Li
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Yuhao Hao
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Kyle Roche
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Guiyuan Chen
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Wen Pan
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Andrew Z Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yuanzeng Min
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China; Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, 230026, China; CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
33
|
He X, Lan H, Jin K, Liu F. Can immunotherapy reinforce chemotherapy efficacy? a new perspective on colorectal cancer treatment. Front Immunol 2023; 14:1237764. [PMID: 37790928 PMCID: PMC10543914 DOI: 10.3389/fimmu.2023.1237764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/25/2023] [Indexed: 10/05/2023] Open
Abstract
As one of the main threats to human life (the fourth most dangerous and prevalent cancer), colorectal cancer affects many people yearly, decreases patients' quality of life, and causes irreparable financial and social damages. In addition, this type of cancer can metastasize and involve the liver in advanced stages. However, current treatments can't completely eradicate this disease. Chemotherapy and subsequent surgery can be mentioned among the current main treatments for this disease. Chemotherapy has many side effects, and regarding the treatment of this type of tumor, chemotherapy can lead to liver damage, such as steatohepatitis, steatosis, and sinus damage. These damages can eventually lead to liver failure and loss of its functions. Therefore, it seems that other treatments can be used in addition to chemotherapy to increase its efficiency and reduce its side effects. Biological therapies and immunotherapy are one of the leading suggestions for combined treatment. Antibodies (immune checkpoint blockers) and cell therapy (DC and CAR-T cells) are among the immune system-based treatments used to treat tumors. Immunotherapy targets various aspects of the tumor that may lead to 1) the recruitment of immune cells, 2) increasing the immunogenicity of tumor cells, and 3) leading to the elimination of inhibitory mechanisms established by the tumor. Therefore, immunotherapy can be used as a complementary treatment along with chemotherapy. This review will discuss different chemotherapy and immunotherapy methods for colorectal cancer. Then we will talk about the studies that have dealt with combined treatment.
Collapse
Affiliation(s)
- Xing He
- Department of Gastroenterology, Jinhua Wenrong Hospital, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Fanlong Liu
- Department of Colorectal Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Mukherjee O, Rakshit S, Shanmugam G, Sarkar K. Role of chemotherapeutic drugs in immunomodulation of cancer. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100068. [PMID: 37692091 PMCID: PMC10491645 DOI: 10.1016/j.crimmu.2023.100068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/12/2023] Open
Abstract
The immune system has a variety of potential effects on a tumor microenvironment and the course of chemotherapy may vary according to that. Anticancer treatments can encourage the release of unwanted signals from senescent tumor cells or the removal of immune-suppressive cells, which can lead to immune system activation. Hence, by inducing an immunological response and conversely making cancer cells more vulnerable to immune attack, chemotherapeutic agents can destroy cancer cells. Furthermore, chemotherapy can activate anticancer immune effectors directly or indirectly by thwarting immunosuppressive pathways. Therefore, in this review, we discuss how chemotherapeutic agents take part in immunomodulation and the molecular mechanisms underlying them. We also focus on the importance of carefully addressing the conflicting effects of chemotherapy on immune responses when developing successful combination treatments based on chemotherapy and immune modulators.
Collapse
Affiliation(s)
- Oishi Mukherjee
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| |
Collapse
|
35
|
Zhang F, Jia Y, Chen F, Zhao Y, Li L, Chang Z. Tumor-targeted bioactive nanoprobes visualizing of hydrogen peroxide for forecasting chemotherapy-exacerbated malignant prognosis. Front Bioeng Biotechnol 2023; 11:1226680. [PMID: 37635993 PMCID: PMC10450909 DOI: 10.3389/fbioe.2023.1226680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction: Fluorescent visualization of hydrogen peroxide in the tumor microenvironment (TME) is conducive to predicting malignant prognosis after chemotherapy. Two photon microscopy has been employed for in vivo hydrogen peroxide detection owing to its advantages of deep penetration and low phototoxicity. Methods: In this study, a two-photon fluorescent probe (TPFP) was protected by mesoporous silica nanoparticles (MSNs) and masked by cloaking the cancer cell membranes (CM), forming a tumor-targeted bioactive nanoprobe, termed MSN@TPFP@CM. Results: This multifunctional nanoprobe allowed for the effective and selective detection of excessive hydrogen peroxide production in chemotherapeutic Etoposide (VP-16)-challenged tumor cells using two-photon microscopy. After specific accumulation in tumors, VP-16-MSN@TPFP@CM monitored tumor-specific hydrogen peroxide levels and revealed a positive correlation between oxidative stress in the TME and chemotherapy-exacerbated malignant prognosis. Discussion: Given the recent translation of fluorescent imaging into early clinical trials and the high biocompatibility of bioactive nanoprobes, our approach may pave the way for specific imaging of oxidative stress in solid tumors after treatment and provide a promising technology for malignant prognosis predictions.
Collapse
Affiliation(s)
- Fan Zhang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, China
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Yong Jia
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Fangman Chen
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, China
| | - Yawei Zhao
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Li Li
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, China
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| | - Zhimin Chang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou, China
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, China
| |
Collapse
|
36
|
Christensen IB, Abrahamsen M, Ribas L, Buch‐Larsen K, Marina D, Andersson M, Larsen S, Schwarz P, Dela F, Gillberg L. Peripheral blood mononuclear cells exhibit increased mitochondrial respiration after adjuvant chemo- and radiotherapy for early breast cancer. Cancer Med 2023; 12:16985-16996. [PMID: 37439084 PMCID: PMC10501284 DOI: 10.1002/cam4.6333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Adjuvant chemo- and radiotherapy cause cellular damage to tumorous and healthy dividing cells. Chemotherapy has been shown to cause mitochondrial respiratory dysfunction in non-tumorous tissues, but the effects on human peripheral blood mononuclear cells (PBMCs) remain unknown. AIM We aimed to investigate mitochondrial respiration of PBMCs before and after adjuvant chemo- and radiotherapy in postmenopausal patients with early breast cancer (EBC) and relate these to metabolic parameters of the patients. METHODS Twenty-three postmenopausal women diagnosed with EBC were examined before and shortly after chemotherapy with (n = 18) or without (n = 5) radiotherapy. Respiration (O2 flux per million PBMCs) was assessed by high-resolution respirometry of intact and permeabilized PBMCs. Clinical metabolic characteristics and mitochondrial DNA (mtDNA) content of PBMCs (mtDN relative to nuclear DNA) were furthermore assessed. RESULTS Respiration of intact and permeabilized PBMCs from EBC patients significantly increased with adjuvant chemo- and radiotherapy (p = 6 × 10-5 and p = 1 × 10-7 , respectively). The oxygen flux attributed to specific mitochondrial complexes and respiratory states increased by 17-43% compared to before therapy initiation. Similarly, PBMC mtDNA content increased by 40% (p = 0.002). Leukocytes (p = 0.0001), hemoglobin (p = 0.0003), and HDL cholesterol (p = 0.003) concentrations decreased whereas triglyceride (p = 0.01) and LDL (p = 0.02) concentrations increased after treatment suggesting a worsened metabolic state. None of the metabolic parameters or the mtDNA content of PBMCs correlated significantly with PBMC respiration. CONCLUSION This study shows that mitochondrial respiration and mtDNA content in circulating PBMCs increase after adjuvant chemo- and radiotherapy in postmenopausal patients with EBC. Besides the increased mtDNA content, a shift in PBMC subpopulation proportions towards cells relying on oxidative phosphorylation, who may be less sensitive to chemotherapy, might influence the increased mitochondrial respiration observed iafter chemotherapy.
Collapse
Affiliation(s)
| | | | - Lucas Ribas
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| | | | - Djordje Marina
- Department of EndocrinologyRigshospitaletCopenhagenDenmark
| | | | - Steen Larsen
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
- Clinical Research CentreMedical University of BialystokBialystokPoland
| | - Peter Schwarz
- Department of EndocrinologyRigshospitaletCopenhagenDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Flemming Dela
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of GeriatricsBispebjerg University HospitalCopenhagenDenmark
| | - Linn Gillberg
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
37
|
McCullum L, Shin J, Xing S, Beekman C, Schuemann J, Hong T, Duda D, Mohan R, Lin SH, Correa-Alfonso CM, Domal S, Withrow J, Bolch W, Paganetti H, Grassberger C. Predicting Severity of Radiation Induced Lymphopenia in Individual Proton Therapy Patients for Varying Dose Rate and Fractionation Using Dynamic 4-Dimensional Blood Flow Simulations. Int J Radiat Oncol Biol Phys 2023; 116:1226-1233. [PMID: 36739919 PMCID: PMC10363211 DOI: 10.1016/j.ijrobp.2023.01.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/11/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
PURPOSE Radiation-induced lymphopenia has gained attention recently as the result of its correlation with survival in a range of indications, particularly when combining radiation therapy (RT) with immunotherapy. The purpose of this study is to use a dynamic blood circulation model combined with observed lymphocyte depletion in patients to derive the in vivo radiosensitivity of circulating lymphocytes and study the effect of RT delivery parameters. METHODS AND MATERIALS We assembled a cohort of 17 patients with hepatocellular carcinoma treated with proton RT alone in 15 fractions (fx) using conventional dose rates (beam-on time [BOT], 120 seconds) for whom weekly absolute lymphocyte counts (ALCs) during RT and follow-up were available. We used HEDOS, a time-dependent, whole-body, blood flow computational framework, in combination with explicit liver blood flow modeling, to calculate the dose volume histograms for circulating lymphocytes for changing BOTs (1 second-300 seconds) and fractionations (5 fx, 15 fx). From this, we used the linear cell survival model and an exponential model to determine patient-specific lymphocyte radiation sensitivity, α, and recovery, σ, respectively. RESULTS The in vivo-derived patient-specific α had a median of 0.65 Gy-1 (range, 0.30-1.38). Decreasing BOT to 1 second led to an increased average end-of-treatment ALC of 27.5%, increasing to 60.3% when combined with the 5-fx regimen. Decreasing to 5 fx at the conventional dose rate led to an increase of 17.0% on average. The benefit of both increasing dose rate and reducing the number of fractions was patient specificࣧpatients with highly sensitive lymphocytes benefited most from decreasing BOT, whereas patients with slow lymphocyte recovery benefited most from the shorter fractionation regimen. CONCLUSIONS We observed that increasing dose rate at the same fractionation reduced ALC depletion more significantly than reducing the number of fractions. High-dose-rates led to an increased sparing of lymphocytes when shortening the fractionation regimen, particularly for patients with radiosensitive lymphocytes at elevated risk.
Collapse
Affiliation(s)
- Lucas McCullum
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Jungwook Shin
- Radiation Epidemiology Branch, National Cancer Institute, Rockville, Maryland
| | - Stella Xing
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Chris Beekman
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Theodore Hong
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dan Duda
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Radhe Mohan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Camilo M Correa-Alfonso
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Sean Domal
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Julia Withrow
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Wesley Bolch
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Clemens Grassberger
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
38
|
Mosele F, Deluche E, Lusque A, Le Bescond L, Filleron T, Pradat Y, Ducoulombier A, Pistilli B, Bachelot T, Viret F, Levy C, Signolle N, Alfaro A, Tran DTN, Garberis IJ, Talbot H, Christodoulidis S, Vakalopoulou M, Droin N, Stourm A, Kobayashi M, Kakegawa T, Lacroix L, Saulnier P, Job B, Deloger M, Jimenez M, Mahier C, Baris V, Laplante P, Kannouche P, Marty V, Lacroix-Triki M, Diéras V, André F. Trastuzumab deruxtecan in metastatic breast cancer with variable HER2 expression: the phase 2 DAISY trial. Nat Med 2023; 29:2110-2120. [PMID: 37488289 PMCID: PMC10427426 DOI: 10.1038/s41591-023-02478-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 06/28/2023] [Indexed: 07/26/2023]
Abstract
The mechanisms of action of and resistance to trastuzumab deruxtecan (T-DXd), an anti-HER2-drug conjugate for breast cancer treatment, remain unclear. The phase 2 DAISY trial evaluated the efficacy of T-DXd in patients with HER2-overexpressing (n = 72, cohort 1), HER2-low (n = 74, cohort 2) and HER2 non-expressing (n = 40, cohort 3) metastatic breast cancer. In the full analysis set population (n = 177), the confirmed objective response rate (primary endpoint) was 70.6% (95% confidence interval (CI) 58.3-81) in cohort 1, 37.5% (95% CI 26.4-49.7) in cohort 2 and 29.7% (95% CI 15.9-47) in cohort 3. The primary endpoint was met in cohorts 1 and 2. Secondary endpoints included safety. No new safety signals were observed. During treatment, HER2-expressing tumors (n = 4) presented strong T-DXd staining. Conversely, HER2 immunohistochemistry 0 samples (n = 3) presented no or very few T-DXd staining (Pearson correlation coefficient r = 0.75, P = 0.053). Among patients with HER2 immunohistochemistry 0 metastatic breast cancer, 5 of 14 (35.7%, 95% CI 12.8-64.9) with ERBB2 expression below the median presented a confirmed objective response as compared to 3 of 10 (30%, 95% CI 6.7-65.2) with ERBB2 expression above the median. Although HER2 expression is a determinant of T-DXd efficacy, our study suggests that additional mechanisms may also be involved. (ClinicalTrials.gov identifier NCT04132960 .).
Collapse
Affiliation(s)
- Fernanda Mosele
- INSERM U981, Gustave Roussy, Villejuif, France
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Elise Deluche
- Department of Medical Oncology, CHU Dupuytren, Limoges, France
| | - Amelie Lusque
- Department of Biostatistics, Institut Claudius-Regaud, IUCT Oncopole, Toulouse, France
| | - Loïc Le Bescond
- INSERM U981, Gustave Roussy, Villejuif, France
- CVN Lab, CentraleSupélec,Université Paris-Saclay, Gif-Sur-Yvette, France
- OPIS, Inria, CentraleSupélec, Université Paris-Saclay, Gif-Sur-Yvette, France
| | - Thomas Filleron
- Department of Biostatistics, Institut Claudius-Regaud, IUCT Oncopole, Toulouse, France
| | - Yoann Pradat
- MICS Lab, CentraleSupélec, Université Paris-Saclay, Gif-Sur-Yvette, France
| | | | - Barbara Pistilli
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Frederic Viret
- Department of Medical Oncology, Centre Paoli Calmettes, Marseille, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Nicolas Signolle
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
| | - Alexia Alfaro
- Imaging and Cytometry Platform, Gustave Roussy, UAR 23/3655, Université Paris-Saclay, Villejuif, France
| | | | | | - Hugues Talbot
- CVN Lab, CentraleSupélec,Université Paris-Saclay, Gif-Sur-Yvette, France
- OPIS, Inria, CentraleSupélec, Université Paris-Saclay, Gif-Sur-Yvette, France
| | | | - Maria Vakalopoulou
- OPIS, Inria, CentraleSupélec, Université Paris-Saclay, Gif-Sur-Yvette, France
- MICS Lab, CentraleSupélec, Université Paris-Saclay, Gif-Sur-Yvette, France
| | - Nathalie Droin
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
| | - Aurelie Stourm
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
| | - Maki Kobayashi
- Translational Research Department, Daiichi Sankyo RD Novare, Tokyo, Japan
| | - Tomoya Kakegawa
- Translational Research Department, Daiichi Sankyo RD Novare, Tokyo, Japan
| | - Ludovic Lacroix
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Patrick Saulnier
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Bastien Job
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
| | - Marc Deloger
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
| | | | | | - Vianney Baris
- UMR9019, CNRS, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Pierre Laplante
- UMR9019, CNRS, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Patricia Kannouche
- UMR9019, CNRS, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Virginie Marty
- AMMICa Platform, INSERM US23, CNRS UAR 3655, AMMICa, Villejuif, France
| | | | - Veronique Diéras
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Fabrice André
- INSERM U981, Gustave Roussy, Villejuif, France.
- Department of Medical Oncology, Gustave Roussy, Villejuif, France.
- Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre, France.
| |
Collapse
|
39
|
Sun XX, Nosrati Z, Ko J, Lee CM, Bennewith KL, Bally MB. Induced Vascular Normalization-Can One Force Tumors to Surrender to a Better Microenvironment? Pharmaceutics 2023; 15:2022. [PMID: 37631236 PMCID: PMC10458586 DOI: 10.3390/pharmaceutics15082022] [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: 05/03/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Immunotherapy has changed the way many cancers are being treated. Researchers in the field of immunotherapy and tumor immunology are investigating similar questions: How can the positive benefits achieved with immunotherapies be enhanced? Can this be achieved through combinations with other agents and if so, which ones? In our view, there is an urgent need to improve immunotherapy to make further gains in the overall survival for those patients that should benefit from immunotherapy. While numerous different approaches are being considered, our team believes that drug delivery methods along with appropriately selected small-molecule drugs and drug candidates could help reach the goal of doubling the overall survival rate that is seen in some patients that are given immunotherapeutics. This review article is prepared to address how immunotherapies should be combined with a second treatment using an approach that could realize therapeutic gains 10 years from now. For context, an overview of immunotherapy and cancer angiogenesis is provided. The major targets in angiogenesis that have modulatory effects on the tumor microenvironment and immune cells are highlighted. A combination approach that, for us, has the greatest potential for success involves treatments that will normalize the tumor's blood vessel structure and alter the immune microenvironment to support the action of immunotherapeutics. So, this is reviewed as well. Our focus is to provide an insight into some strategies that will engender vascular normalization that may be better than previously described approaches. The potential for drug delivery systems to promote tumor blood vessel normalization is considered.
Collapse
Affiliation(s)
- Xu Xin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Zeynab Nosrati
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Janell Ko
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
| | - Che-Min Lee
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kevin L. Bennewith
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
40
|
Marcelin HN, Dasse RS, Yeboah RO, Tariam AD, Kagambega AGZ, Oseni AM, Kouassi YKK, Bilé MA, Toure M, Thakar M, Adoubi I, Kizub D. Circulating natural killer cells and their association with breast cancer and its clinico-pathological characteristics. Ecancermedicalscience 2023; 17:1567. [PMID: 37533944 PMCID: PMC10393315 DOI: 10.3332/ecancer.2023.1567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Indexed: 08/04/2023] Open
Abstract
Purpose Natural killer (NK) cells play a critical role in cancer immunosurveillance and hold promise as both therapies and prognostic markers in advanced disease. We explore factors that may influence NK cell concentration in the peripheral blood of women with breast cancer in Côte d'Ivoire compared to healthy controls and implications for future research in our context. Methods In this cross-sectional case-control study, blood samples were taken from 30 women diagnosed with breast cancer within 6 months of diagnosis and fifteen healthy women at University Teaching Hospital [Centre Hospitalier Universitaire (CHU)] Treichville in Abidjan, Côte d'Ivoire, from March to September 2018. The blood draw could take place at any time following diagnosis and through treatment. Demographic and clinical data were collected. NK cells were isolated, stained, analysed and counted using the flow cytometer at the Department of Immunology at CHU of Cocody. All p-values were two-sided. Results Mean age among 30 women with breast cancer was 49 years old compared to 45 years old for 15 controls (p = 0.41). Among 30 women with breast cancer, 4 (13.3%) had Stage 2 disease, 14 (46.7 %) at Stage 3, and 12 (40%) at Stage 4. Fourteen (46.7%) had breast cancer that was hormone receptor-positive (HR+) HER2-negative, 10 (33.3%) had triple-negative cancer, three (10.0%) had HR+HER2+ disease, and three (10.0%) HR-HER2+ cancer. NK cell concentration was not associated with cancer diagnosis, age, cancer stage, subtype, or type of treatment patients received (p > 0.05). Conclusion Although we did not find an association between NK cell concentration, cancer characteristics or treatment, our results be limited by the small sample size and timing of blood draw. Our next steps include a larger study to explore circulating NK cells prior to any treatment and NK cell infiltration within breast cancer tumour and correlating this with response to treatment and prognosis.
Collapse
Affiliation(s)
- Homian N’da Marcelin
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Romuald S Dasse
- Department of Immunology, Centre Hospitalier Universitaire de Cocody, BP V 3, Abidjan, Côte d’Ivoire
| | - Richard O Yeboah
- Department of Immunology, Centre Hospitalier Universitaire de Cocody, BP V 3, Abidjan, Côte d’Ivoire
| | - Agnès D Tariam
- Department of Immunology, Centre Hospitalier Universitaire de Cocody, BP V 3, Abidjan, Côte d’Ivoire
| | - Arsène G Z Kagambega
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Akandji M Oseni
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Y K K Kouassi
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Michel A Bilé
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Moctar Toure
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Monica Thakar
- The Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Innocent Adoubi
- Department of Oncology, Centre Hospitalier Universitaire de Treichville, 7XVV+5P4, Abidjan, Côte d’Ivoire
| | - Darya Kizub
- University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
41
|
Page DB, Pucilowska J, Chun B, Kim I, Sanchez K, Moxon N, Mellinger S, Wu Y, Koguchi Y, Conrad V, Redmond WL, Martel M, Sun Z, Campbell MB, Conlin A, Acheson A, Basho R, McAndrew P, El-Masry M, Park D, Bennetts L, Seitz RS, Nielsen TJ, McGregor K, Rajamanickam V, Bernard B, Urba WJ, McArthur HL. A phase Ib trial of pembrolizumab plus paclitaxel or flat-dose capecitabine in 1st/2nd line metastatic triple-negative breast cancer. NPJ Breast Cancer 2023; 9:53. [PMID: 37344474 DOI: 10.1038/s41523-023-00541-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/21/2023] [Indexed: 06/23/2023] Open
Abstract
Chemoimmunotherapy with anti-programmed cell death 1/ligand 1 and cytotoxic chemotherapy is a promising therapeutic modality for women with triple-negative breast cancer, but questions remain regarding optimal chemotherapy backbone and biomarkers for patient selection. We report final outcomes from a phase Ib trial evaluating pembrolizumab (200 mg IV every 3 weeks) with either weekly paclitaxel (80 mg/m2 weekly) or flat-dose capecitabine (2000 mg orally twice daily for 7 days of every 14-day cycle) in the 1st/2nd line setting. The primary endpoint is safety (receipt of 2 cycles without grade III/IV toxicities requiring discontinuation or ≥21-day delays). The secondary endpoint is efficacy (week 12 objective response). Exploratory aims are to characterize immunologic effects of treatment over time, and to evaluate novel biomarkers. The trial demonstrates that both regimens meet the pre-specified safety endpoint (paclitaxel: 87%; capecitabine: 100%). Objective response rate is 29% for pembrolizumab/paclitaxel (n = 4/13, 95% CI: 10-61%) and 43% for pembrolizumab/capecitabine (n = 6/14, 95% CI: 18-71%). Partial responses are observed in two subjects with chemo-refractory metaplastic carcinoma (both in capecitabine arm). Both regimens are associated with significant peripheral leukocyte contraction over time. Response is associated with clinical PD-L1 score, non-receipt of prior chemotherapy, and the H&E stromal tumor-infiltrating lymphocyte score, but also by a novel 27 gene IO score and spatial biomarkers (lymphocyte spatial skewness). In conclusion, pembrolizumab with paclitaxel or capecitabine is safe and clinically active. Both regimens are lymphodepleting, highlighting the competing immunostimulatory versus lymphotoxic effects of cytotoxic chemotherapy. Further exploration of the IO score and spatial TIL biomarkers is warranted. The clinical trial registration is NCT02734290.
Collapse
Affiliation(s)
- David B Page
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| | - Joanna Pucilowska
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Brie Chun
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Isaac Kim
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Katherine Sanchez
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Nicole Moxon
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Staci Mellinger
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Yaping Wu
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Yoshinobu Koguchi
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Valerie Conrad
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Maritza Martel
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Zhaoyu Sun
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Mary B Campbell
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Alison Conlin
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Anupama Acheson
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Reva Basho
- Cedars Sinai Medical Center, Los Angeles, CA, USA
- Ellison Institute for Transformative Medicine, Los Angeles, CA, USA
| | | | | | - Dorothy Park
- Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Laura Bennetts
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | | | | | | | - Brady Bernard
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Walter J Urba
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Heather L McArthur
- Cedars Sinai Medical Center, Los Angeles, CA, USA
- UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
42
|
Zou X, Lin X, Cheng H, Chen Y, Wang R, Ma M, Liu Y, Dai Z, Tasiheng Y, Yan Y, Hou Q, Ding F, Chen H, Yu X, Wang X, Liu C. Characterization of intratumoral tertiary lymphoid structures in pancreatic ductal adenocarcinoma: cellular properties and prognostic significance. J Immunother Cancer 2023; 11:e006698. [PMID: 37364934 PMCID: PMC10410893 DOI: 10.1136/jitc-2023-006698] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Tumor-associated tertiary lymphoid structures (TLSs) are functional immune-responsive niches that are not fully understood in pancreatic ductal adenocarcinoma (PDAC). METHODS Fluorescent multiplex immunohistochemistry was performed on sequential sections of surgically resected tumor tissues from 380 PDAC patients without preoperative treatment (surgery alone (SA)) and 136 patients pretreated with neoadjuvant treatment (NAT). Multispectral images were processed via machine learning and image processing platforms, inForm V.2.4 and HALO V.3.2; TLS regions were segmented, and the cells were identified and quantified. The cellular composition and immunological properties of TLSs and their adjacent tissues in PDAC were scored and compared, and their association with prognosis was further examined. RESULTS Intratumoral TLSs were identified in 21.1% (80/380) of patients in the SA group and 15.4% (21/136) of patients in the NAT group. In the SA group, the presence of intratumoral TLSs was significantly associated with improved overall survival (OS) and progression-free survival. The existence of intratumoral TLSs was correlated with elevated levels of infiltrating CD8+T, CD4+T, B cells and activated immune cells in adjacent tissues. A nomogram model was generated with TLS presence as a variable, which successfully predicted PDAC patient OS in an external validation cohort (n=123). In the NAT group, samples exhibited a lower proportion of B cells and a higher proportion of regulatory T cells within intratumoral TLSs. Additionally, these TLSs were smaller in size, with a lower overall maturation level and reduced immune cell activation, and the prognostic value of TLS presence was insignificant in the NAT cohort. CONCLUSION Our study systematically revealed the cellular properties and prognostic values of intratumoral TLSs in PDAC and described the potential impact of NAT on TLS development and function.
Collapse
Affiliation(s)
- Xuan Zou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yusheng Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Ruijie Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Mingjian Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhengjie Dai
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yesboli Tasiheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Yan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Qinqin Hou
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fei Ding
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Huan Chen
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xu Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Radiation Oncology, Cancer Research Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
43
|
Tadesse FA, Leminie AA. Effects of Adriamycin-Cytoxan chemotherapy on hematological and electrolyte parameters among breast cancer patients. Front Oncol 2023; 13:1103013. [PMID: 37205205 PMCID: PMC10185890 DOI: 10.3389/fonc.2023.1103013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/03/2023] [Indexed: 05/21/2023] Open
Abstract
Background Adriamycin-Cytoxan (AC) is a common chemotherapy treatment for breast cancer (BC) patients. Its electrolyte and hematological adverse effects have not been addressed adequately. Objective This study aimed to assess the effect of AC on hematological and electrolyte parameters among BC patients. Methods A hospital-based comparative cross-sectional study design was conducted from March to November 2022. Randomly selected AC-treated (n=100) and untreated (n=100) patients were included. Structured questionnaire and medical records were used to collect sociodemographic data. Anthropometric parameters, hematological indices, and serum electrolytes were measured. Cobas Integra 400+and SYSMEX-XT-4000i were used to analyze serum electrolytes and hematological indices respectively. The data were analyzed using SPSS version 25. Independent t-test and chi-square test were used. p-value <0.05 was considered statistically significant. Results AC-treated patients' mean total white blood cell (TWBC), neutrophil (NE), lymphocyte (LY), red blood cell (RBC), hemoglobin (Hgb), hematocrit (HCT), and sodium(Na+) values were significantly reduced (p<0.05) than patients with no treatment. However, mean eosinophils (EO), platelet (PLT) counts, red cell distribution-width (RDW), potassium (K+), and plateletcrit (PCT values were significantly increased (p<0.05). Conclusion The majority of blood cells and serum sodium were affected by AC treatment. Incorporating these parameters in the routine analysis and further studies on the detailed mechanism of action of this drug is required.
Collapse
Affiliation(s)
| | - Abebaye Aragaw Leminie
- Department of Physiology, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
44
|
Emens LA, Loi S. Immunotherapy Approaches for Breast Cancer Patients in 2023. Cold Spring Harb Perspect Med 2023; 13:13/4/a041332. [PMID: 37011999 PMCID: PMC10071416 DOI: 10.1101/cshperspect.a041332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Immunotherapy, particularly agents targeting the immunoregulatory PD-1/PD-L1 axis, harnesses the power of the immune system to treat cancer, with unique potential for a durable treatment effect due to immunologic memory. The PD-1 inhibitor pembrolizumab combined with neoadjuvant chemotherapy followed by adjuvant pembrolizumab improves event-free survival and is a new standard of care for high-risk, early-stage triple-negative breast cancer (TNBC), regardless of tumor PD-L1 expression. For metastatic TNBC, pembrolizumab combined with chemotherapy is a new standard of care for the first-line therapy of PD-L1+ metastatic TNBC, with improvement in overall survival. The PD-L1 inhibitor atezolizumab combined with nab-paclitaxel is also approved outside the United States for the first-line treatment of metastatic PD-L1+ TNBC. Current research focuses on refining the use of immunotherapy in TNBC by defining informative predictive biomarkers, developing immunotherapy in early and advanced HER2-driven and luminal breast cancers, and overcoming primary and secondary resistance to immunotherapy through unique immune-based strategies.
Collapse
Affiliation(s)
- Leisha A Emens
- Department of Medicine, University of Pittsburgh/UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232, USA
- Ankyra Therapeutics, Boston, Massachusetts 02116, USA
| | - Sherene Loi
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| |
Collapse
|
45
|
Papp KA, Melosky B, Sehdev S, Hotte SJ, Beecker JR, Kirchhof MG, Turchin I, Dutz JP, Gooderham MJ, Gniadecki R, Hong CH, Lambert J, Lynde CW, Prajapati VH, Vender RB. Use of Systemic Therapies for Treatment of Psoriasis in Patients with a History of Treated Solid Tumours: Inference-Based Guidance from a Multidisciplinary Expert Panel. Dermatol Ther (Heidelb) 2023; 13:867-889. [PMID: 36929121 PMCID: PMC10060504 DOI: 10.1007/s13555-023-00905-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Patients with treated solid tumours (TSTs) are a highly heterogeneous population at an increased risk for malignancy compared with the general population. When treating psoriasis in patients with a history of TSTs, clinicians are concerned about the immunosuppressive nature of psoriasis therapies, the possibility of augmenting cancer recurrence/progression, and infectious complications. No direct, high-level evidence exists to address these concerns. OBJECTIVES We aim to provide a structured framework supporting healthcare professional and patient discussions on the risks and benefits of systemic psoriasis therapy in patients with previously TSTs. Our goal was to address the clinically important question, "In patients with TSTs, does therapy with systemic agents used for psoriasis increase the risk of malignancy or malignancy recurrence?" METHODS We implemented an inference-based approach relying on indirect evidence when direct clinical trial and real-world data were absent. We reviewed indirect evidence supporting inferences on the status of immune function in patients with TSTs. Recommendations on systemic psoriasis therapies in patients with TSTs were derived using an inferential heuristic. RESULTS We identified five indirect indicators of iatrogenic immunosuppression informed by largely independent bodies of evidence: (1) overall survival, (2) rate of malignancies with psoriasis and systemic psoriasis therapies, (3) rate of infections with psoriasis and systemic psoriasis therapies, (4) common disease biochemical pathways for solid tumours and systemic psoriasis therapies, and (5) solid organ transplant outcomes. On the basis of review of the totality of this data, we provided inference-based conclusions and ascribed level of support for each statement. CONCLUSIONS Prior to considering new therapies for psoriasis, an understanding of cancer prognosis should be addressed. Patients with TSTs and a good cancer prognosis will have similar outcomes to non-TST patients when treated with systemic psoriasis therapies. For patients with TSTs and a poor cancer prognosis, the quality-of-life benefits of treating psoriasis may outweigh the theoretical risks.
Collapse
Affiliation(s)
- Kim A Papp
- Probity Medical Research Inc., Waterloo, ON, Canada.
- Alliance Clinical Research, Waterloo, ON, Canada.
| | - Barbara Melosky
- Medical Oncology, BC Cancer Vancouver Centre, Vancouver, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sandeep Sehdev
- Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Sebastien J Hotte
- Juravinski Cancer Centre, Hamilton, ON, Canada
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Jennifer R Beecker
- Probity Medical Research Inc., Waterloo, ON, Canada
- University of Ottawa, Ottawa, ON, Canada
- Division of Dermatology, The Ottawa Hospital, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mark G Kirchhof
- University of Ottawa, Ottawa, ON, Canada
- Division of Dermatology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Irina Turchin
- Probity Medical Research Inc., Waterloo, ON, Canada
- Brunswick Dermatology Centre, Fredericton, NB, Canada
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jan P Dutz
- Skin Care Centre, Vancouver, BC, Canada
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Melinda J Gooderham
- Probity Medical Research Inc., Waterloo, ON, Canada
- SKiN Centre for Dermatology, Peterborough, ON, Canada
| | - Robert Gniadecki
- Division of Dermatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Chih-Ho Hong
- Probity Medical Research Inc., Waterloo, ON, Canada
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
- Dr. Chih-ho Hong Medical Inc., Surrey, BC, Canada
| | - Jo Lambert
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
- Dermatology Research Unit, Ghent University, Ghent, Belgium
| | - Charles W Lynde
- Probity Medical Research Inc., Waterloo, ON, Canada
- Lynde Institute for Dermatology, Markham, ON, Canada
- Division of Dermatology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Vimal H Prajapati
- Probity Medical Research Inc., Waterloo, ON, Canada
- Division of Dermatology, Department of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Community Pediatrics, Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Section of Pediatric Rheumatology, Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Dermatology Research Institute, Calgary, AB, Canada
- Skin Health & Wellness Centre, Calgary, AB, Canada
| | - Ronald B Vender
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Dermatrials Research Inc., Hamilton, ON, Canada
| |
Collapse
|
46
|
Zhao X, Liu Z, Chen T. Potential Role of Vaginal Microbiota in Ovarian Cancer Carcinogenesis, Progression and Treatment. Pharmaceutics 2023; 15:pharmaceutics15030948. [PMID: 36986809 PMCID: PMC10056320 DOI: 10.3390/pharmaceutics15030948] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Ovarian cancer represents one of the most challenging gynecologic cancers which still has numerous unknowns on the underlying pathogenesis. In addition to the verified contributors such as genomic predisposition and medical history in the carcinogenesis, emerging evidence points out the potential role of vaginal microbiota in ovarian cancer. Recent studies have underlined the presence of vaginal microbial dysbiosis in cancer cases. Increasing research also indicates the potential correlations between vaginal microbes and cancer carcinogenesis, progression and treatment. Currently, compared with other gynecologic cancers, reports on the roles of vaginal microbiota in ovarian cancer remain scarce and fragmentary. Therefore, in this review, we summarize the roles of vaginal microbiota in various gynecologic diseases, particularly focusing on the potential mechanisms and possible applications of vaginal microbiota in ovarian cancer, giving insight into the involvement of vaginal microbiota in gynecologic cancer treatment.
Collapse
Affiliation(s)
- Xiumiao Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Correspondence: (Z.L.); (T.C.)
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
- Correspondence: (Z.L.); (T.C.)
| |
Collapse
|
47
|
Wu X, Tan Y, Zhang J, Cui R, Liao C, Zhang S. Nanodrug constructed using dietary antioxidants for immunotherapy of metastatic tumors. J Mater Chem B 2023; 11:2916-2926. [PMID: 36892505 DOI: 10.1039/d2tb02773a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Immunogenic cell death (ICD) induced by reactive oxygen species (ROS) represents a particular form of tumor cell death for approaching the problem of low immunogenicity of tumors in immunotherapy, while the oxidative damage to normal cells of current ICD inducers hinders their clinical application. Herein, a new ICD inducer VC@cLAV constructed solely by dietary antioxidants, lipoic acid (LA) and vitamin C (VC), is developed, which could promote heavy intracellular ROS production in cancer cells for ICD induction while acting as an anti-oxidant in non-cancer cells for cytoprotection, and thus hold high biosafety. In vitro studies show that VC@cLAV induced a release of antigens and a maturation rate of DCs up to 56.5%, approaching the positive control (58.4%). In vivo combined with αPD-1, VC@cLAV showed excellent antitumor activity against both primary and distant metastatic tumors with an inhibition rate of 84.8% and 79.0% compared to 14.2% and 10.0% in the αPD-1 alone group. Notably, VC@cLAV established a long-term antitumor immune memory effect against tumor rechallenging. This study not only presents a new kind of ICD inducer but also provides an impetus for the development of dietary antioxidant-based cancer drugs.
Collapse
Affiliation(s)
- Xiao Wu
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Yifeng Tan
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Jing Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Rong Cui
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Chunyan Liao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Shiyong Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
48
|
Utilizing chemotherapy-induced tumor RNA nanoparticles to improve cancer chemoimmunotherapy. Acta Biomater 2023; 158:698-707. [PMID: 36563773 DOI: 10.1016/j.actbio.2022.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Chemotherapy has become a popular combination strategy to improve the response rate of immunotherapy since certain chemotherapeutic drugs kill tumor cells by an immunogenic cell death (ICD) pathway, which activates antitumor immune responses. Unfortunately, the synergistic effect of chemoimmunotherapy can be impaired due to the toxicities of chemotherapeutic agent-induced lymphatic depletion and immunosuppression. In this study, we present an approach to improve immunotherapy by using tumor RNA nanoparticles (RNA-NPs) where RNA is directly extracted from chemotherapy-treated cancer cells and then condensed by protamine via electrostatic interactions to form complexes. Such RNA-NPs can be effectively taken up by dendritic cells (DCs) in the draining lymph nodes after subcutaneous injection. Compared with noninduced tumor RNA nanoparticles (N-RNA-NPs), chemotherapy-induced tumor RNA nanoparticles (C-RNA-NPs) can significantly promote DC maturation and stimulate a stronger immune response against established CT-26 colon carcinoma. Besides, C-RNA-NPs can improve the efficacy of immune checkpoint blockade (ICB) therapy by facilitating the infiltration of intratumoral T cells and increasing the ratio of CD8+ T cells to regulatory T cells (Tregs). More importantly, the synergistic effect of chemoimmunotherapy is also enhanced by treatment with C-RNA-NPs. STATEMENT OF SIGNIFICANCE: Although immune checkpoint blockade therapy has been demonstrated to be effective in some advanced cancers, the low response rate has significantly limited its clinical application. To address this issue, a new strategy for improving cancer immunotherapy using chemotherapy-induced tumor RNA nanoparticles (C-RNA-NPs) is developed in this work. The proposed C-RNA-NPs could be captured by dendritic cells, which were then stimulated to the maturation status to initiate an anticancer immune response. Furthermore, the response rate to immunotherapy was significantly increased by promoting intratumoral T-cell infiltration and elevating the intratumoral ratio of CD8+ T cells to regulatory T cells after treatment with C-RNA-NPs. Therefore, C-RNA-NPs have the potential to improve cancer immunotherapy.
Collapse
|
49
|
Rizvi N, Ademuyiwa FO, Cao ZA, Chen HX, Ferris RL, Goldberg SB, Hellmann MD, Mehra R, Rhee I, Park JC, Kluger H, Tawbi H, Sullivan RJ. Society for Immunotherapy of Cancer (SITC) consensus definitions for resistance to combinations of immune checkpoint inhibitors with chemotherapy. J Immunother Cancer 2023; 11:e005920. [PMID: 36918220 PMCID: PMC10016262 DOI: 10.1136/jitc-2022-005920] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 03/15/2023] Open
Abstract
Although immunotherapy can offer profound clinical benefit for patients with a variety of difficult-to-treat cancers, many tumors either do not respond to upfront treatment with immune checkpoint inhibitors (ICIs) or progressive/recurrent disease occurs after an interval of initial control. Improved response rates have been demonstrated with the addition of ICIs to cytotoxic therapies, leading to approvals from the US Food and Drug Administration and regulatory agencies in other countries for ICI-chemotherapy combinations in a number of solid tumor indications, including breast, head and neck, gastric, and lung cancer. Designing trials for patients with tumors that do not respond or stop responding to treatment with immunotherapy combinations, however, is challenging without uniform definitions of resistance. Previously, the Society for Immunotherapy of Cancer (SITC) published consensus definitions for resistance to single-agent anti-programmed cell death protein 1 (PD-1). To provide guidance for clinical trial design and to support analyses of emerging molecular and cellular data surrounding mechanisms of resistance to ICI-based combinations, SITC convened a follow-up workshop in 2021 to develop consensus definitions for resistance to multiagent ICI combinations. This manuscript reports the consensus clinical definitions for combinations of ICIs and chemotherapies. Definitions for resistance to ICIs in combination with targeted therapies and with other ICIs will be published in companion volumes to this paper.
Collapse
Affiliation(s)
| | | | | | - Helen X Chen
- National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | - Ranee Mehra
- University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ina Rhee
- Genentech, South San Francisco, California, USA
| | - Jong Chul Park
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Hussein Tawbi
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
50
|
Li M, Quintana A, Alberts E, Hung MS, Boulat V, Ripoll MM, Grigoriadis A. B Cells in Breast Cancer Pathology. Cancers (Basel) 2023; 15:1517. [PMID: 36900307 PMCID: PMC10000926 DOI: 10.3390/cancers15051517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
B cells have recently become a focus in breast cancer pathology due to their influence on tumour regression, prognosis, and response to treatment, besides their contribution to antigen presentation, immunoglobulin production, and regulation of adaptive responses. As our understanding of diverse B cell subsets in eliciting both pro- and anti-inflammatory responses in breast cancer patients increases, it has become pertinent to address the molecular and clinical relevance of these immune cell populations within the tumour microenvironment (TME). At the primary tumour site, B cells are either found spatially dispersed or aggregated in so-called tertiary lymphoid structures (TLS). In axillary lymph nodes (LNs), B cell populations, amongst a plethora of activities, undergo germinal centre reactions to ensure humoral immunity. With the recent approval for the addition of immunotherapeutic drugs as a treatment option in the early and metastatic settings for triple-negative breast cancer (TNBC) patients, B cell populations or TLS may resemble valuable biomarkers for immunotherapy responses in certain breast cancer subgroups. New technologies such as spatially defined sequencing techniques, multiplex imaging, and digital technologies have further deciphered the diversity of B cells and the morphological structures in which they appear in the tumour and LNs. Thus, in this review, we comprehensively summarise the current knowledge of B cells in breast cancer. In addition, we provide a user-friendly single-cell RNA-sequencing platform, called "B singLe cEll rna-Seq browSer" (BLESS) platform, with a focus on the B cells in breast cancer patients to interrogate the latest publicly available single-cell RNA-sequencing data collected from diverse breast cancer studies. Finally, we explore their clinical relevance as biomarkers or molecular targets for future interventions.
Collapse
Affiliation(s)
- Mengyuan Li
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
| | | | - Elena Alberts
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Miu Shing Hung
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
| | - Victoire Boulat
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Mercè Martí Ripoll
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Biosensing and Bioanalysis Group, Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Anita Grigoriadis
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- Breast Cancer Now Unit, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
| |
Collapse
|