1
|
Ippolito A, Wang H, Zhang Y, Vakil V, Popel AS. Virtual clinical trials via a QSP immuno-oncology model to simulate the response to a conditionally activated PD-L1 targeting antibody in NSCLC. J Pharmacokinet Pharmacodyn 2024; 51:747-757. [PMID: 38858306 PMCID: PMC11579200 DOI: 10.1007/s10928-024-09928-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/19/2024] [Indexed: 06/12/2024]
Abstract
Recently, immunotherapies for antitumoral response have adopted conditionally activated molecules with the objective of reducing systemic toxicity. Amongst these are conditionally activated antibodies, such as PROBODY® activatable therapeutics (Pb-Tx), engineered to be proteolytically activated by proteases found locally in the tumor microenvironment (TME). These PROBODY® therapeutics molecules have shown potential as PD-L1 checkpoint inhibitors in several cancer types, including both effectiveness and locality of action of the molecule as shown by several clinical trials and imaging studies. Here, we perform an exploratory study using our recently published quantitative systems pharmacology model, previously validated for triple-negative breast cancer (TNBC), to computationally predict the effectiveness and targeting specificity of a PROBODY® therapeutics drug compared to the non-modified antibody. We begin with the analysis of anti-PD-L1 immunotherapy in non-small cell lung cancer (NSCLC). As a first contribution, we have improved previous virtual patient selection methods using the omics data provided by the iAtlas database portal compared to methods previously published in literature. Furthermore, our results suggest that masking an antibody maintains its efficacy while improving the localization of active therapeutic in the TME. Additionally, we generalize the model by evaluating the dependence of the response to the tumor mutational burden, independently of cancer type, as well as to other key biomarkers, such as CD8/Treg Tcell and M1/M2 macrophage ratio. While our results are obtained from simulations on NSCLC, our findings are generalizable to other cancer types and suggest that an effective and highly selective conditionally activated PROBODY® therapeutics molecule is a feasible option.
Collapse
Affiliation(s)
- Alberto Ippolito
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vahideh Vakil
- Clinical and Quantitative Pharmacology, CytomX Therapeutics, Inc., South San Francisco, CA, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Oncology, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
2
|
Aliyeva T, Aktas BY, Gundogdu F, Chalabiyev E, Arik Z, Usubutun A. The predictive role of PD-L1 expression and CD8 + TIL levels in determining the neoadjuvant chemotherapy response in advanced ovarian cancer. J Ovarian Res 2024; 17:234. [PMID: 39580458 PMCID: PMC11585239 DOI: 10.1186/s13048-024-01533-x] [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: 03/09/2024] [Accepted: 10/08/2024] [Indexed: 11/25/2024] Open
Abstract
OBJECTIVE To analyze how the PD-L1 expression and CD8 + tumor infiltrating lymphocyte (TIL) levels in biopsy samples before neoadjuvant chemotherapy (NACT) can predict chemotherapy response score and survival for advanced high-grade serous ovarian cancer (HGSC). METHODS We retrospectively analyzed 45 patients with advanced epithelial ovarian cancer between 2010 and 2018, who had received at least three cycles of NACT. PD-L1 expression and CD8 + TIL levels were evaluated by immunohistochemical staining in the pre-NAC tumor samples from which the patients had been diagnosed. The post-NACT tissue samples taken during interval debulking surgery (IDS) were used to evaluate the chemotherapy response score (CRS). RESULTS Among all the patients, CRS 1 (no response) was found in 8 patients, CRS 2 (partial response) in 28 patients, and CRS 3 (complete response) in 9 patients. A total of 20 (44.4%) patients had high intratumoral CD8 + TILs (iCD8 + TILs) levels, and 35 (77.8%) patients had high expression stromal CD8 + TILs (sCD8 + TILs). No statistically significant relationship was found between high and low expression of i/s CD8 + TILs levels with PFS and CRS. The study found that 33 (73.3%) patients had high levels of stromal PD-L1 (sPD-L1) expression and 28 (62.2%) patients had high levels of intratumoral PD-L1 (iPD-L1) expression. In the iPD-L1 group, patients with low expression had a PFS of 28 months, whereas those with high expression had a PFS of 17 months (p = 0.028). Among the patients with high iPD-L1 expression, 23 (82.1%) patients showed CRS2, 4 (14.3%) showed CRS3, and only 1 (3.6%) showed CRS1 (p < 0.001). However, high or low expression sPD-L1 did not significantly affect PFS and CRS (p = 0.928 and p = 0.305; respectively). CONCLUSIONS We found that iPD-L1 expression levels in diagnostic biopsy in ovarian cancer can predict the chemotherapy response score in interval debulking surgery.
Collapse
Affiliation(s)
- T Aliyeva
- Department of Internal Medicine, Hacettepe University School of Medicine, Ankara, Turkey.
| | - B Y Aktas
- Division of Medical Oncology, Hacettepe University School of Medicine, Ankara, Turkey
| | - F Gundogdu
- Department of Pathology, Hacettepe University School of Medicine, Ankara, Turkey
| | - E Chalabiyev
- Division of Medical Oncology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Z Arik
- Division of Medical Oncology, Hacettepe University School of Medicine, Ankara, Turkey
| | - A Usubutun
- Department of Pathology, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
3
|
Dharmapuri S, Cabal R, Akturk G, Ioannou G, Ozbey S, Paulsen J, Raina S, Ang C, Sarpel U, Sung MW, Kozuch P, Schwartz ME, Cohen DJ, Gnjatic S, Pintova S. Multiplexed immunohistochemical analysis of the immune microenvironment of biliary tract cancers pre- & post-neoadjuvant chemotherapy: case series. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:78. [PMID: 39118963 PMCID: PMC11304425 DOI: 10.21037/atm-23-1928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/17/2024] [Indexed: 08/10/2024]
Abstract
Background Neoadjuvant chemotherapy (NACT) is increasingly being used in the management of locally advanced biliary tract cancer (BTC). The evidence suggests a contributing role of tumor infiltrating immune cells in the prognosis and response. We set out to characterize immune modulation of tumor immune microenvironment in BTC following NACT. Case Description Patients with BTC who underwent diagnostic biopsy, then NACT then resection between 2014-2018 were identified. Multiplexed immunohistochemical consecutive staining on single slide (MICSSS) analysis was performed with a series of immune markers to characterize T-cells, immune checkpoints etc. on pre- & post-NACT tumor tissue. Density was calculated for each marker. The final analysis included five patients. Median age was 48 (range, 41-56) years, with 4 female, 4 intrahepatic cholangiocarcinoma and 1 gallbladder. All patients received gemcitabine/cisplatin as NACT (median of 5 cycles). Median time from diagnosis to surgery was 4.3 (range, 1.4-7.8) months. All patients were mismatch repair proficient (pMMR). NACT on average produced a depletion of all immune markers. Given small sample size, each patient was considered their own control and changes in mean cell densities post-NACT were calculated. Patient #2 with a 40-fold increase in PD-L1 expression & 5-fold decrease in CD8:FOXP3 ratio after NACT notably had the shortest disease-free interval (DFI). Patient #3 with the longest DFI had the largest increase in CD8:FOXP3 by about 8-fold with a decrease in PD-L1. Conclusions Preliminary results suggest NACT may differentially modulate various compartments of the immune tumor contexture despite overall cell depletion. Future studies should focus on strategies to expand immune modulation of tumor microenvironment, including immune-oncology agents to augment the effects of chemotherapy.
Collapse
Affiliation(s)
- Sirish Dharmapuri
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| | - Rafael Cabal
- Division of Molecular and Cell-Based Medicine, Department of Pathology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Guray Akturk
- Division of Molecular and Cell-Based Medicine, Department of Pathology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Giorgio Ioannou
- Division of Molecular and Cell-Based Medicine, Department of Pathology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Sinem Ozbey
- Division of Molecular and Cell-Based Medicine, Department of Pathology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - John Paulsen
- Division of Molecular and Cell-Based Medicine, Department of Pathology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Sheen Raina
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| | - Celina Ang
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| | - Umut Sarpel
- Division of Surgical Oncology, Department of Surgery, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Max W. Sung
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| | - Peter Kozuch
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| | - Myron E. Schwartz
- Division of Surgical Oncology, Department of Surgery, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Deirdre Jill Cohen
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| | - Sacha Gnjatic
- Division of Molecular and Cell-Based Medicine, Department of Pathology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Sofya Pintova
- Division of Medical Oncology, Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai West, Tisch Cancer Institute, New York, NY, USA
| |
Collapse
|
4
|
Ulevicius J, Jasukaitiene A, Bartkeviciene A, Dambrauskas Z, Gulbinas A, Urboniene D, Paskauskas S. Preoperative Immune Cell Dysregulation Accompanies Ovarian Cancer Patients into the Postoperative Period. Int J Mol Sci 2024; 25:7087. [PMID: 39000195 PMCID: PMC11240929 DOI: 10.3390/ijms25137087] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Ovarian cancer (OC) poses a significant global health challenge with high mortality rates, emphasizing the need for improved treatment strategies. The immune system's role in OC progression and treatment response is increasingly recognized, particularly regarding peripheral blood mononuclear cells (PBMCs) and cytokine production. This study aimed to investigate PBMC subpopulations (T and B lymphocytes, natural killer cells, monocytes) and cytokine production, specifically interleukin-1 beta (IL-1β), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-12 (IL-12), and tumor necrosis factor alpha (TNFα), in monocytes of OC patients both preoperatively and during the early postoperative period. Thirteen OC patients and 23 controls were enrolled. Preoperatively, OC patients exhibited changes in PBMC subpopulations, including decreased cytotoxic T cells, increased M2 monocytes, and the disbalance of monocyte cytokine production. These alterations persisted after surgery with subtle additional changes observed in PBMC subpopulations and cytokine expression in monocytes. Considering the pivotal role of these altered cells and cytokines in OC progression, our findings suggest that OC patients experience an enhanced pro-tumorigenic environment, which persists into the early postoperative period. These findings highlight the impact of surgery on the complex interaction between the immune system and OC progression. Further investigation is needed to clarify the underlying mechanisms during this early postoperative period, which may hold potential for interventions aimed at improving OC management.
Collapse
Affiliation(s)
- Jonas Ulevicius
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| | - Aldona Jasukaitiene
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| | - Arenida Bartkeviciene
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| | - Zilvinas Dambrauskas
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| | - Antanas Gulbinas
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| | - Daiva Urboniene
- Department of Laboratory Medicine, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| | - Saulius Paskauskas
- Department of Obstetrics and Gynecology, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| |
Collapse
|
5
|
Mazzoccoli L, Liu B. Dendritic Cells in Shaping Anti-Tumor T Cell Response. Cancers (Basel) 2024; 16:2211. [PMID: 38927916 PMCID: PMC11201542 DOI: 10.3390/cancers16122211] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Among professional antigen-presenting cells, dendritic cells (DCs) orchestrate innate and adaptive immunity and play a pivotal role in anti-tumor immunity. DCs are a heterogeneous population with varying functions in the tumor microenvironment (TME). Tumor-associated DCs differentiate developmentally and functionally into three main subsets: conventional DCs (cDCs), plasmacytoid DCs (pDCs), and monocyte-derived DCs (MoDCs). There are two major subsets of cDCs in TME, cDC1 and cDC2. cDC1 is critical for cross-presenting tumor antigens to activate cytotoxic CD8+ T cells and is also required for priming earlier CD4+ T cells in certain solid tumors. cDC2 is vital for priming anti-tumor CD4+ T cells in multiple tumor models. pDC is a unique subset of DCs and produces type I IFN through TLR7 and TLR9. Studies have shown that pDCs are related to immunosuppression in the TME through the secretion of immunosuppressive cytokines and by promoting regulatory T cells. MoDCs differentiate separately from monocytes in response to inflammatory cues and infection. Also, MoDCs can cross-prime CD8+ T cells. In this review, we summarize the subsets and functions of DCs. We also discuss the role of different DC subsets in shaping T cell immunity in TME and targeting DCs for potential immunotherapeutic benefits against cancer.
Collapse
Affiliation(s)
- Luciano Mazzoccoli
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Cunha D, Neves M, Silva D, Silvestre AR, Nunes PB, Arrobas F, Ribot JC, Ferreira F, Moita LF, Soares-de-Almeida L, Silva JM, Filipe P, Ferreira J. Tumor-Infiltrating T Cells in Skin Basal Cell Carcinomas and Squamous Cell Carcinomas: Global Th1 Preponderance with Th17 Enrichment-A Cross-Sectional Study. Cells 2024; 13:964. [PMID: 38891095 PMCID: PMC11172364 DOI: 10.3390/cells13110964] [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: 05/04/2024] [Revised: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Basal cell carcinomas (BCCs) and squamous cell carcinomas (SCCs) are high-incidence, non-melanoma skin cancers (NMSCs). The success of immune-targeted therapies in advanced NMSCs led us to anticipate that NMSCs harbored significant populations of tumor-infiltrating lymphocytes with potential anti-tumor activity. The main aim of this study was to characterize T cells infiltrating NMSCs. Flow cytometry and immunohistochemistry were used to assess, respectively, the proportions and densities of T cell subpopulations in BCCs (n = 118), SCCs (n = 33), and normal skin (NS, n = 30). CD8+ T cells, CD4+ T cell subsets, namely, Th1, Th2, Th17, Th9, and regulatory T cells (Tregs), CD8+ and CD4+ memory T cells, and γδ T cells were compared between NMSCs and NS samples. Remarkably, both BCCs and SCCs featured a significantly higher Th1/Th2 ratio (~four-fold) and an enrichment for Th17 cells. NMSCs also showed a significant enrichment for IFN-γ-producing CD8+T cells, and a depletion of γδ T cells. Using immunohistochemistry, NMSCs featured denser T cell infiltrates (CD4+, CD8+, and Tregs) than NS. Overall, these data favor a Th1-predominant response in BCCs and SCCs, providing support for immune-based treatments in NMSCs. Th17-mediated inflammation may play a role in the progression of NMSCs and thus become a potential therapeutic target in NMSCs.
Collapse
Affiliation(s)
- Daniela Cunha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Centro de Dermatologia, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal
- Dermatology Unit, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Marco Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
| | - Daniela Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
| | - Ana Rita Silvestre
- Serviço de Anatomia Patológica, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal (P.B.N.)
| | - Paula Borralho Nunes
- Serviço de Anatomia Patológica, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal (P.B.N.)
- Instituto de Anatomia Patológica, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Fernando Arrobas
- Datamedica, Biostatistics Services and Consulting, 2610-008 Amadora, Portugal
| | - Julie C. Ribot
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
| | - Fernando Ferreira
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Luís F. Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Luís Soares-de-Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Serviço de Dermatologia, Centro Hospitalar Universitário Lisboa Norte EPE, 1649-028 Lisbon, Portugal
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - João Maia Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Centro de Dermatologia, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal
- Serviço de Dermatologia, Centro Hospitalar Universitário Lisboa Norte EPE, 1649-028 Lisbon, Portugal
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Paulo Filipe
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Serviço de Dermatologia, Centro Hospitalar Universitário Lisboa Norte EPE, 1649-028 Lisbon, Portugal
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - João Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| |
Collapse
|
7
|
Kment J, Newsted D, Young S, Vermeulen MC, Laight BJ, Greer PA, Lan Y, Craig AW. Blockade of TGF-β and PD-L1 by bintrafusp alfa promotes survival in preclinical ovarian cancer models by promoting T effector and NK cell responses. Br J Cancer 2024; 130:2003-2015. [PMID: 38622286 PMCID: PMC11183086 DOI: 10.1038/s41416-024-02677-9] [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: 10/03/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Failure of immunotherapy in high-grade serous ovarian cancer (HGSC) may be due to high levels of transforming growth factor-β (TGF-β) in ascites or tumour immune microenvironment (TIME). Here, we test whether coordinated blockade of TGF-β and PD-L1 with bintrafusp alfa (BA) can provoke anti-tumour immune responses in preclinical HGSC models. METHODS BA is a first-in-class bifunctional inhibitor of TGF-β and PD-L1, and was tested for effects on overall survival and altered TIME in syngeneic HGSC models. RESULTS Using a mouse ID8-derived HGSC syngeneic model with IFNγ-inducible PD-L1 expression, BA treatments significantly reduced ascites development and tumour burden. BA treatments depleted TGF-β and VEGF in ascites, and skewed the TIME towards cytotoxicity compared to control. In the BR5 HGSC syngeneic model, BA treatments increased tumour-infiltrating CD8 T cells with effector memory and cytotoxic markers, as well as cytolytic NK cells. Extended BA treatments in the BR5 model produced ∼50% BA-cured mice that were protected from re-challenge. These BA-cured mice had increased peritoneal T-effector memory and NK cells compared to controls. CONCLUSIONS Our preclinical studies of BA in advanced ovarian cancer models support further testing of BA as an improved immunotherapy option for patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Jacob Kment
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Daniel Newsted
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Stephanie Young
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Michael C Vermeulen
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Brian J Laight
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Peter A Greer
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Yan Lan
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Andrew W Craig
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
8
|
Occhiuto CJ, Liby KT. KEAP1-Mutant Lung Cancers Weaken Anti-Tumor Immunity and Promote an M2-like Macrophage Phenotype. Int J Mol Sci 2024; 25:3510. [PMID: 38542481 PMCID: PMC10970780 DOI: 10.3390/ijms25063510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
Considerable advances have been made in lung cancer therapies, but there is still an unmet clinical need to improve survival for lung cancer patients. Immunotherapies have improved survival, although only 20-30% of patients respond to these treatments. Interestingly, cancers with mutations in Kelch-like ECH-associated protein 1 (KEAP1), the negative regulator of the nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor, are resistant to immune checkpoint inhibition and correlate with decreased lymphoid cell infiltration. NRF2 is known for promoting an anti-inflammatory phenotype when activated in immune cells, but the study of NRF2 activation in cancer cells has not been adequately assessed. The objective of this study was to determine how lung cancer cells with constitutive NRF2 activity interact with the immune microenvironment to promote cancer progression. To assess, we generated CRISPR-edited mouse lung cancer cell lines by knocking out the KEAP1 or NFE2L2 genes and utilized a publicly available single-cell dataset through the Gene Expression Omnibus to investigate tumor/immune cell interactions. We show here that KEAP1-mutant cancers promote immunosuppression of the tumor microenvironment. Our data suggest KEAP1 deletion is sufficient to alter the secretion of cytokines, increase expression of immune checkpoint markers on cancer cells, and alter recruitment and differential polarization of immunosuppressive macrophages that ultimately lead to T-cell suppression.
Collapse
Affiliation(s)
- Christopher J. Occhiuto
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen T. Liby
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Peng X, Lu X, Yang D, Liu J, Wu H, Peng H, Zhang Y. A novel CD8+ T cell-related gene signature as a prognostic biomarker in hepatocellular carcinoma. Medicine (Baltimore) 2024; 103:e37496. [PMID: 38489709 PMCID: PMC10939595 DOI: 10.1097/md.0000000000037496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/16/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
CD8+ T cells have great roles in tumor suppression and elimination of various tumors including hepatocellular carcinoma (HCC). Nonetheless, potential prognostic roles of CD8+ T cell-related genes (CD8Gs) in HCC remains unknown. In our study, 416 CD8Gs were identified in HCC, which were enriched in inflammatory and immune signaling pathways. Using The Cancer Genome Atlas dataset, a 5-CD8Gs risk model (KLRB1, FYN, IL2RG, FCER1G, and DGKZ) was constructed, which was verified in International Cancer Genome Consortium and gene expression omnibus datasets. Furthermore, we found that overall survival was independently correlated with the CD8Gs signature, and it was associated with immune- and cancer-related signaling pathways and immune cells infiltration. Finally, drug sensitivity data indicated that 10 chemotherapeutic drugs held promise as therapeutics for HCC patients with high-risk. In conclusion, multi-databases analysis showed that 5-CD8Gs and their signature could be an indicator to predict candidate drugs for HCC therapy.
Collapse
Affiliation(s)
- Xiaozhen Peng
- School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua, China
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, China
| | - Xingjun Lu
- School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Daqing Yang
- School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Jinyan Liu
- Hunan Normal University, Changsha, China
| | - Honglin Wu
- School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Hong Peng
- Medical School, Huanghe Science & Technology College, Zhengzhou, China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Han KH, Kim CH, Kim SH, Lee CH, Park M, Bui VD, Duong VH, Kwon S, Ha M, Kang H, Park JH. Immunogenic Extracellular Vesicles Derived from Endoplasmic Reticulum-Stressed Tumor Cells: Implications as the Therapeutic Cancer Vaccine. ACS NANO 2024; 18:199-209. [PMID: 38109681 DOI: 10.1021/acsnano.3c05645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Tumor-derived extracellular vesicles (TDEs) have potential for therapeutic cancer vaccine applications since they innately possess tumor-associated antigens, mediate antigen presentation, and can incorporate immune adjuvants for enhanced vaccine efficacy. However, the original TDEs also contain immune-suppressive proteins. To address this, we proposed a simple yet powerful preconditioning method to improve the overall immunogenicity of the TDEs. This approach involved inducing endoplasmic reticulum (ER) stress on parental tumor cells via N-glycosylation inhibition with tunicamycin. The generated immunogenic TDEs (iTDEs) contained down-regulated immunosuppressive proteins and up-regulated immune adjuvants, effectively activating dendritic cells (DCs) in vitro. Furthermore, in vivo evidence from a tumor-bearing mouse model showed that iTDEs activated DCs, enabling cytotoxic T lymphocytes (CTLs) to target tumors, and eventually established a systemic antitumor immune response. Additionally, iTDEs significantly delayed tumor recurrence in a postsurgery model compared with control groups. These findings highlight the immense potential of our strategy for utilizing TDEs to develop effective cancer vaccines.
Collapse
Affiliation(s)
- Kyung Hee Han
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - So Hee Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Chang Hyun Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Minsung Park
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351 Republic of Korea
| | - Van Dat Bui
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Van Hieu Duong
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Seunglee Kwon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Minji Ha
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Heegun Kang
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351 Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| |
Collapse
|
11
|
Kumar S, Acharya S, Karthikeyan M, Biswas P, Kumari S. Limitations and potential of immunotherapy in ovarian cancer. Front Immunol 2024; 14:1292166. [PMID: 38264664 PMCID: PMC10803592 DOI: 10.3389/fimmu.2023.1292166] [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: 09/11/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Ovarian cancer (OC) is the third most common gynecological cancer and alone has an emergence rate of approximately 308,069 cases worldwide (2020) with dire survival rates. To put it into perspective, the mortality rate of OC is three times higher than that of breast cancer and it is predicted to only increase significantly by 2040. The primary reasons for such a high rate are that the physical symptoms of OC are detectable only during the advanced phase of the disease when resistance to chemotherapies is high and around 80% of the patients that do indeed respond to chemotherapy initially, show a poor prognosis subsequently. This highlights a pressing need to develop new and effective therapies to tackle advanced OC to improve prognosis and patient survival. A major advance in this direction is the emergence of combination immunotherapeutic methods to boost CD8+ T cell function to tackle OC. In this perspective, we discuss our view of the current state of some of the combination immunotherapies in the treatment of advanced OC, their limitations, and potential approaches toward a safer and more effective response.
Collapse
Affiliation(s)
| | | | | | | | - Sudha Kumari
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| |
Collapse
|
12
|
Miceska S, Skof E, Bucek S, Kuhar CG, Gasljevic G, Smrkolj S, Prevodnik VK. The prognostic significance of tumor-immune microenvironment in ascites of patients with high-grade serous carcinoma. Radiol Oncol 2023; 57:493-506. [PMID: 38038414 PMCID: PMC10690755 DOI: 10.2478/raon-2023-0046] [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: 06/27/2023] [Accepted: 08/13/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND High-grade serous carcinoma (HGSC) is often associated with ascites at presentation. Our objective was to quantify immune cells (ICs) in ascites prior to any treatment was given and evaluate their impact on progression-free survival (PFS) and overall survival (OS). PATIENTS AND METHODS Forty-seven patients with primary HGSC and ascites were included. Flow-cytometric analysis was performed to detect percentages of CD3+ T cells (CD4+, CD8+, Tregs, and NKT cells), B cells, NK cells (CD56brightCD16- and CD56dimCD16+ subsets), macrophages and dendritic cells (DCs). Furthermore, CD103 expression was analyzed on T cells and their subsets, while PD-1 and PD-L1 expression on all ICs. Cut-off of low and high percentages of ICs was determined by the median of variables, and correlation with PFS and OS was calculated. RESULTS CD3+ cells were the predominant ICs (median 51%), while the presence of other ICs was much lower (median ≤10%). CD103+ expression was mostly present on CD8+, and not CD4+ cells. PD-1 was mainly expressed on CD3+ T cells (median 20%), lower expression was observed on other ICs (median ≤10%). PD-L1 expression was not detected. High percentages of CD103+CD3+ T cells, PD-1+ Tregs, CD56brightCD16- NK cells, and DCs correlated with prolonged PFS and OS, while high percentages of CD8+ cells, macrophages, and PD-1+CD56brightCD16- NK cells, along with low percentages of CD4+ cells, correlated with better OS only. DCs were the only independent prognostic marker among all ICs. CONCLUSIONS Our results highlight the potential of ascites tumor-immune microenvironment to provide additional prognostic information for HGSC patients. However, a larger patient cohort and longer follow-up are needed to confirm our findings.
Collapse
Affiliation(s)
- Simona Miceska
- Department of Cytopathology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Erik Skof
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Simon Bucek
- Department of Cytopathology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Cvetka Grasic Kuhar
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Gorana Gasljevic
- Department of Pathology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Spela Smrkolj
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Division of Gynaecology and Obstetrics, University Medical Centre, Ljubljana, Slovenia
| | - Veronika Kloboves Prevodnik
- Department of Cytopathology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
13
|
Robertson SE, Yasukawa M, Marchion DC, Xiong Y, Naqvi SMH, Gheit T, Tommasino M, Wenham RM, Giuliano AR, Lancaster JM, Shahzad MMK. Prevalence of viral DNA in high-grade serous epithelial ovarian cancer and correlation with clinical outcomes. PLoS One 2023; 18:e0294448. [PMID: 38039311 PMCID: PMC10691703 DOI: 10.1371/journal.pone.0294448] [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: 07/08/2023] [Accepted: 11/01/2023] [Indexed: 12/03/2023] Open
Abstract
INTRODUCTION Currently 11 infectious agents are classified as carcinogenic but the role of infectious agents on outcomes of epithelial ovarian cancer is largely unknown. OBJECTIVE To explore the association between infectious agents and ovarian cancer, we investigated the prevalence of viral DNA in primary ovarian cancer tumors and its association with clinical outcomes. METHODS Archived tumors from 98 patients diagnosed with high-grade serous epithelial ovarian cancer were collected between 1/1/1994 and 12/31/2010. After DNA extraction, Luminex technology was utilized to identify polymerase chain reaction-amplified viral DNA for 113 specific viruses. Demographic data and disease characteristics were summarized using descriptive statistics. We used logistic regression and Cox proportional hazards model to assess associations between tumor viral status and disease outcome and between tumor viral presence and overall survival (OS), respectively. RESULTS Forty-six cases (45.9%) contained at least one virus. Six highly prevalent viruses were associated with clinical outcomes and considered viruses of interest (VOI; Epstein-Barr virus 1, Merkel cell polyomavirus, human herpes virus 6b, and human papillomaviruses 4, 16, and 23). Factors independently associated with OS were presence of VOI (HR 4.11, P = 0.0001) and platinum sensitivity (HR 0.21, P<0.0001). Median OS was significantly decreased when tumors showed VOI versus not having these viruses (22 vs 44 months, P<0.0001). Women <70 year old with VOI in tumors had significantly lower median OS versus age-matched women without VOI (20 vs 57 months, P = 0.0006); however, among women ≥70 years old, there was no difference in OS by tumor virus status. CONCLUSIONS The presence of a VOI was significantly associated with a lower OS. These findings may have implications for clinical management of ovarian cancer but require additional studies.
Collapse
Affiliation(s)
- Sharon E. Robertson
- Department of Gynecology Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Maya Yasukawa
- Department of Gynecology Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Douglas C. Marchion
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Yin Xiong
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Syeda Mahrukh Hussnain Naqvi
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Tarik Gheit
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Massimo Tommasino
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Robert M. Wenham
- Department of Gynecology Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, United States of America
| | - Anna R. Giuliano
- Risk Assessment, Detection and Intervention Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Johnathan M. Lancaster
- Department of Gynecology Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, United States of America
| | - Mian M. K. Shahzad
- Department of Gynecology Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, United States of America
| |
Collapse
|
14
|
Farokhi Boroujeni S, Rodriguez G, Galpin K, Yakubovich E, Murshed H, Ibrahim D, Asif S, Vanderhyden BC. BRCA1 and BRCA2 deficient tumour models generate distinct ovarian tumour microenvironments and differential responses to therapy. J Ovarian Res 2023; 16:231. [PMID: 38017453 PMCID: PMC10683289 DOI: 10.1186/s13048-023-01313-z] [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: 08/18/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
Clinical trials are currently exploring combinations of PARP inhibitors and immunotherapies for the treatment of ovarian cancer, but their effects on the ovarian tumour microenvironment (TME) remain unclear. Here, we investigate how olaparib, PD-L1 monoclonal antibodies, and their combination can influence TME composition and survival of tumour-bearing mice. We further explored how BRCA deficiencies can influence the response to therapy. Olaparib and combination therapies similarly improved the median survival of Brca1- and Brca2-deficient tumour-bearing mice. Anti-PD-L1 monotherapy improved the survival of mice with Brca1-null tumours, but not Brca2-null tumours. A detailed analysis of the TME revealed that olaparib monotherapy resulted in a large number of immunosuppressive and immunomodulatory effects in the more inflamed Brca1-deficient TME but not Brca2-deficient tumours. Anti-PD-L1 treatment was mostly immunosuppressive, resulting in a systemic reduction of cytokines and a compensatory increase in PD-L1 expression. The results of the combination therapy generally resembled the effects of one or both of the monotherapies, along with unique changes observed in certain immune populations. In-silico analysis of RNA-seq data also revealed numerous differences between Brca-deficient tumour models, such as the expression of genes involved in inflammation, angiogenesis and PD-L1 expression. In summary, these findings shed light on the influence of novel therapeutics and BRCA mutations on the ovarian TME.
Collapse
Affiliation(s)
- Salar Farokhi Boroujeni
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Galaxia Rodriguez
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Kristianne Galpin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Edward Yakubovich
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Humaira Murshed
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Dalia Ibrahim
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Sara Asif
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
15
|
Yang R, Yang M, Wu Z, Liu B, Zheng M, Lu L, Wu S. Tespa1 deficiency reduces the antitumour immune response by decreasing CD8 +T cell activity in a mouse Lewis lung cancer model. Int Immunopharmacol 2023; 124:110865. [PMID: 37660596 DOI: 10.1016/j.intimp.2023.110865] [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: 06/05/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Thymocyte-expressed, positive selection-associated 1 (Tespa1) is a key molecule in T-cell development and has been linked to immune diseases. However, its role in antitumour CD8+T cell immunity remains unclear. Here, we demonstrated that Tespa1 plays an important role in antitumour CD8+T cell immunity. First, compared with wild-type (WT) mice, Lewis lung cancer cells grew faster in Tespa1 knockout (Tespa1-/-) mice, with reduced apoptosis, and decreased CD8+T cells in peripheral blood and tumor tissues. Second, the proportion of CD8+T and Th1 cells in the splenocytes of Tespa1-/- mice was lower than that in WT mice. Third, Tespa1-/- CD8+ tumor-infiltrating lymphocytes (TILs) showed weakened proliferation, invasion, cytotoxicity, and protein expression of IL-2 signalling pathway components compared to WT CD8+TILs. Furthermore, PD-1 expression in CD8+TILs was higher in Tespa1-/- than in WT mice. Lastly, CD8+TILs in WT mice improved the antitumour ability of Tespa1-/- mice. In conclusion, these findings suggest that Tespa1 plays a critical role in the tumor immune system by regulating CD8+T cells.
Collapse
Affiliation(s)
- Ruhui Yang
- School of Medicine and Pharmaceutical Engineering, Taizhou Vocational and Technical College, Taizhou 318000, China; Department of Pharmacology, Lishui University School of Medicine, Lishui 323000, China
| | - Mingyue Yang
- The First Clinical Department, China Medical University, Shenyang 110122, China
| | - Zehua Wu
- Faculty of Science and Engineering, University of Nottingham, Ningbo, 315000, China
| | - Bingjin Liu
- School of Medicine and Pharmaceutical Engineering, Taizhou Vocational and Technical College, Taizhou, 318000, China
| | - Mingzhu Zheng
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Linrong Lu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Songquan Wu
- Department of Immunology, Lishui University School of Medicine, Lishui 323000, China.
| |
Collapse
|
16
|
Sicard G, Protzenko D, Giacometti S, Barlési F, Ciccolini J, Fanciullino R. Harnessing tumor immunity with cytotoxics: T cells monitoring in mice bearing lung tumors treated with anti-VEGF and pemetrexed-cisplatin doublet. Br J Cancer 2023; 129:1373-1382. [PMID: 37524968 PMCID: PMC10628115 DOI: 10.1038/s41416-023-02350-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/19/2023] [Accepted: 06/27/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Successful immunotherapy is restricted to some cancers only, and combinatorial strategies with other drugs could help to improve their efficacy. Here, we monitor T cells in NSCLC model after treatment with cytotoxics (CT) and anti-VEGF drugs, to understand when immune checkpoint inhibitors should be best associated next. METHODS In vivo study was performed on BALB/c mice grafted with KLN205 cells. Eight treatments were tested including control, cisplatin and pemetrexed as low (LD CT) and full (MTD CT) dose as single agents, flat dose anti-VEGF and the association anti-VEGF + CT. Full immunomonitoring was performed by flow cytometry on tumor, spleen and blood over 3 weeks. RESULTS Immunomodulatory effect was dependent upon both treatments and time. In tumors, combination groups shown numerical lower Treg cells on Day 21. In spleen, anti-VEGF and LD CT group shown higher CD8/Treg ratio on Day 7; on Day 14, higher T CD4 were observed in both combination groups. Finally, in blood, Tregs were lower and CD8/Treg ratio higher, on Day 14 in both combination groups. On Day 21, CD4 and CD8 T cells were higher in the anti-VEGF + MTD CT group. CONCLUSIONS Anti-VEGF associated to CT triggers notable increase in CD8/Tregs ratio. Regarding the scheduling, a two-week delay after using anti-VEGF and CT could be the best sequence to optimize antitumor efficacy.
Collapse
Affiliation(s)
- G Sicard
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - D Protzenko
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - S Giacometti
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - F Barlési
- School of Medicine, Aix Marseille University, 13007, Marseille, France
- Gustave Roussy Institute, 94800, Villejuif, France
| | - J Ciccolini
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France.
| | - R Fanciullino
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| |
Collapse
|
17
|
Barna AJ, Herold Z, Acs M, Bazsa S, Gajdacsi J, Garay TM, Herold M, Madaras L, Muhl D, Nagy A, Szasz AM, Dank M. High Tumor-Infiltrating Lymphocyte Count Is Associated with Distinct Gene Expression Profile and Longer Patient Survival in Advanced Ovarian Cancer. Int J Mol Sci 2023; 24:13684. [PMID: 37761986 PMCID: PMC10530512 DOI: 10.3390/ijms241813684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/26/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer-related immunity plays a significant role in the outcome of ovarian cancer, but the exact mechanisms are not fully explored. A retrospective, real-life observational study was conducted including 57 advanced ovarian cancer patients. Immunohistochemistry for CD4+, CD8+, and CD45+ was used for assessing tumor-infiltrating immune cells. Furthermore, an immune-related gene expression assay was performed on 12-10 samples from patients with less than and more than 1-year overall survival (OS), respectively. A higher number of CD4+ (p = 0.0028) and CD45+ (p = 0.0221) immune cells within the tumor microenvironment were associated with longer OS of patients. In a multivariate setting, higher CD4+ T cell infiltration predicted longer OS (p = 0.0392). Twenty-three differentially expressed genes-involved in antigen presentation, costimulatory signaling, matrix remodeling, metastasis formation, and myeloid cell activity-were found when comparing the prognostic groups. It was found that tumor-infiltrating immune cell counts are associated with peculiar gene expression patterns and bear prognostic information in ovarian cancer. SOX11 expression emerged and was validated as a predictive marker for OS.
Collapse
Affiliation(s)
- Andras Jozsef Barna
- Department of Obstetrics and Gynecology, Saint Pantaleon Hospital, H-2400 Dunaujvaros, Hungary
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Zoltan Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Miklos Acs
- Department of Surgery, University Hospital, D-93053 Regensburg, Germany
| | - Sandor Bazsa
- Department of Obstetrics and Gynecology, Saint Pantaleon Hospital, H-2400 Dunaujvaros, Hungary
| | - Jozsef Gajdacsi
- Directorate General of Medical Quality Assurance, Semmelweis University, H-1085 Budapest, Hungary
| | - Tamas Marton Garay
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Faculty of Information Technology and Bionics, Pazmany Peter Catholic University, H-1083 Budapest, Hungary
| | - Magdolna Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Department of Internal Medicine and Hematology, Semmelweis University, H-1088 Budapest, Hungary
| | - Lilla Madaras
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, H-1091 Budapest, Hungary
| | - Dorottya Muhl
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Akos Nagy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Attila Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Magdolna Dank
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| |
Collapse
|
18
|
Pizarro D, Romero I, Pérez-Mies B, Redondo A, Caniego-Casas T, Carretero-Barrio I, Cristóbal E, Gutiérrez-Pecharromán A, Santaballa A, D'Angelo E, Hardisson D, Vieites B, Matías-Guiu X, Estévez P, Guerra E, Prat J, Poveda A, López-Guerrero JA, Palacios J. The Prognostic Significance of Tumor-Infiltrating Lymphocytes, PD-L1, BRCA Mutation Status and Tumor Mutational Burden in Early-Stage High-Grade Serous Ovarian Carcinoma-A Study by the Spanish Group for Ovarian Cancer Research (GEICO). Int J Mol Sci 2023; 24:11183. [PMID: 37446361 DOI: 10.3390/ijms241311183] [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: 05/09/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Early stages are under-represented in studies on the molecular and immune features of high-grade serous ovarian carcinoma (HGSOC), and specific studies focused on early-stage HGSOC are required for a better prognostic stratification and to personalize chemotherapy. The aim of this study was to determine the prognostic significance of CD8+ and CD4+ tumor-infiltrating lymphocytes (TILs), tumoral cell PD-L1 expression, BRCA mutational status and tumor mutation burden (TMB) in early-stage HGSOC. A retrospective study was performed on stage I and II HGSOC from the Molecular Reclassification of Early Stages of Ovarian Cancer (RECLAMO) cohort from the Spanish Group of Ovarian Cancer Research (GEICO). Centralized histological typing was performed based on morphological and immunohistochemical features. Intraepithelial (i) and stromal (s) CD8+ and CD4+ T cells and PD-L1 were evaluated on tissue microarrays by immunohistochemistry. BRCA1 and BRCA2 mutation status and TMB were analyzed in tumor DNA using next-generation sequencing. The study included 124 tumors. High iCD8+ (>20 TILs/core), low/intermediate CD4+ (<20 TILs/core) and high CD8+/CD4+ ratio (>35/core) were associated with favorable outcomes. Tumor cell PD-L1 expression (TPS ≥ 1) was present in only 8% of tumors. In total, 11 (16%) and 6 (9%) out of 69 HGSOC tested carried pathogenic or likely pathogenic BRCA1 or BRCA2 mutations, respectively. Median TMB of 40 tumors analyzed was 5.04 mutations/Mb and only 6 tumors had 10 or more mutations/Mb. BRCA status and TMB were not associated with TILs or prognosis. When compared with studies on advanced HGSOC, our results suggested that prognostic variables differed according to stage and that more studies focused on early stages of HGSOC are needed to better stratify these tumors.
Collapse
Affiliation(s)
- David Pizarro
- Pathology Department, University Hospital Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
| | - Ignacio Romero
- Instituto Valenciano de Oncología, 46009 Valencia, Spain
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
| | - Belén Pérez-Mies
- Pathology Department, University Hospital Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Faculty of Medicine, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Andrés Redondo
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
- Oncology Department, University Hospital La Paz, IdiPAZ, 28046 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Tamara Caniego-Casas
- Pathology Department, University Hospital Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Irene Carretero-Barrio
- Pathology Department, University Hospital Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Faculty of Medicine, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Eva Cristóbal
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Ana Santaballa
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
- Oncology Department, University Hospital La Fe, 46026 Valencia, Spain
| | - Emanuela D'Angelo
- Department of Medical, Oral, and Biotechnological Sciences, University "G.D'Annunzio" of Chieti-Pescara, 66013 Chieti, Italy
| | - David Hardisson
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
- Pathology Department, University Hospital La Paz, 28046 Madrid, Spain
| | - Begoña Vieites
- Pathology Department, University Hospital Virgen del Rocío, 41013 Sevilla, Spain
| | - Xavier Matías-Guiu
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Pathology and Medical Oncology Departments, Hospital Universitari Arnau de Vilanova, IRBLLEIDA, University of Lleida, 25003 Lleida, Spain
- Pathology Department, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, 08007 Barcelona, Spain
| | - Purificación Estévez
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
- Oncology Department, University Hospital Virgen del Rocío, 41013 Sevilla, Spain
- Seville Biomedical Research Institute (IBIS), 41013 Sevilla, Spain
| | - Eva Guerra
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
- Oncology Department, University Hospital Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
| | - Jaime Prat
- Pathology Department, Emeritus Faculty, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Andrés Poveda
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
- Initia Oncología, Hospital Quironsalud Valencia, 46010 Valencia, Spain
| | - José Antonio López-Guerrero
- Instituto Valenciano de Oncología, 46009 Valencia, Spain
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
| | - José Palacios
- Pathology Department, University Hospital Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
- Spanish Group for Investigation on Ovarian Cancer (GEICO), 28003 Madrid, Spain
- Biomedical Research Network in Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Faculty of Medicine, University of Alcalá, 28801 Alcalá de Henares, Spain
| |
Collapse
|
19
|
Dong T, Shah JR, Phung AT, Larson C, Sanchez AB, Aisagbonhi O, Blair SL, Oronsky B, Trogler WC, Reid T, Kummel AC. A Local and Abscopal Effect Observed with Liposomal Encapsulation of Intratumorally Injected Oncolytic Adenoviral Therapy. Cancers (Basel) 2023; 15:3157. [PMID: 37370769 PMCID: PMC10296131 DOI: 10.3390/cancers15123157] [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: 02/16/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
This study evaluated the in vivo therapeutic efficacy of oncolytic serotype 5 adenovirus TAV255 in CAR-deficient tumors. In vitro experiments were performed with cell lines that expressed different levels of CAR (HEK293, A549, CT26, 4T1, and MCF-7). Low CAR cells, such as CT26, were poorly transduced by Ad in vitro unless the adenovirus was encapsulated in liposomes. However, the CT26 tumor in an immune-competent mouse model responded to the unencapsulated TAV255; 33% of the tumors were induced into complete remission, and mice with complete remission rejected the rechallenge with cancer cell injection. Encapsulation of TAV255 improves its therapeutic efficacy by transducing more CT26 cells, as expected from in vitro results. In a bilateral tumor model, nonencapsulated TAV255 reduced the growth rate of the locally treated tumors but had no effect on the growth rate of the distant tumor site. Conversely, encapsulated TAV255-infected CT26 induced a delayed growth rate of both the primary injected tumor and the distant tumor, consistent with a robust immune response. In vivo, intratumorally injected unencapsulated adenoviruses infect CAR-negative cells with only limited efficiency. However, unencapsulated adenoviruses robustly inhibit the growth of CAR-deficient tumors, an effect that constitutes an 'in situ vaccination' by stimulating cytotoxic T cells.
Collapse
Affiliation(s)
- Tao Dong
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Jaimin R. Shah
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Materials Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Abraham T. Phung
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | - Omonigho Aisagbonhi
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Sarah L. Blair
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
| | | | - William C. Trogler
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony Reid
- EpicentRx, Inc., La Jolla, CA 92037, USA
| | - Andrew C. Kummel
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
20
|
Sun G, Yang Z, Fang K, Xiong Y, Tu S, Yi S, Xiao W. Distribution characteristics and clinical significance of infiltrating T cells in the tumor microenvironment of pancreatic cancer. Oncol Lett 2023; 25:261. [PMID: 37205920 PMCID: PMC10189847 DOI: 10.3892/ol.2023.13847] [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: 07/28/2022] [Accepted: 03/24/2023] [Indexed: 05/21/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are important components of the tumor microenvironment (TME). However, the distribution characteristics of TILs and their significance in pancreatic cancer (PC) remain largely unexplored. The levels of TILs, including the total number of T cells, cluster of differentiation (CD)4+ T cells, CD8+ cytotoxic T lymphocytes (CTLs), regulatory T-cells (Tregs), programmed cell death protein 1+ T cells and programmed cell death ligand 1 (PD-L1)+ T cells, in the TME of patients with PC were detected using multiple fluorescence immunohistochemistry. The associations between the number of TILs and the clinicopathological characteristics were investigated using χ2 tests. In addition, Kaplan-Meier survival and Cox regression analyses were used to assess the prognostic value of these TIL types. Compared with paracancerous tissues, in PC tissues, the proportions of total T cells, CD4+ T cells and CD8+ CTLs were markedly decreased, while those of Tregs and PD-L1+ T cells were significantly increased. The levels of CD4+ T cell and CD8+ CTL infiltrates were inversely associated with tumor differentiation. Higher infiltrates of Tregs and PD-L1+ T cells were closely associated with advanced N and TNM stages. It is important to note that the infiltrates of total T cells, CD4+ T cells, Tregs and PD-L1+ T cells in the TME were independent risk factors for the prognosis of PC. PC was characterized by an immunosuppressive TME with a decrease in the number of CD4+ T cells and CD8+ CTLs, and an increase in the number of Tregs and PD-L1+ T cells. Overall, the number of total T cells, CD4+ T cells, Tregs and PD-L1+ T cells in the TME was a potential predictive marker for the prognosis of PC.
Collapse
Affiliation(s)
- Gen Sun
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhengjiang Yang
- Department of General Surgery, The Affiliated Hospital of Jiujiang College, Jiujiang, Jiangxi 332001, P.R. China
| | - Kang Fang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuanpeng Xiong
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shuju Tu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Siqing Yi
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Weidong Xiao
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Correspondence to: Dr Weidong Xiao, Department of General Surgery, The First Affiliated Hospital of Nanchang University, 17 Yongwaizhengjie, Nanchang, Jiangxi 330006, P.R. China, E-mail:
| |
Collapse
|
21
|
Lim JU, Lee E, Lee SY, Cho HJ, Ahn DH, Hwang Y, Choi JY, Yeo CD, Park CK, Kim SJ. Current literature review on the tumor immune micro-environment, its heterogeneity and future perspectives in treatment of advanced non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:857-876. [PMID: 37197639 PMCID: PMC10183402 DOI: 10.21037/tlcr-22-633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/07/2023] [Indexed: 03/21/2023]
Abstract
Background and Objective Immune checkpoint inhibitors (ICI) were a major clinical advancement that provided an opportunity to improve the prognosis of patients with non-small cell lung cancer (NSCLC). However, programmed death-ligand-1 (PD-L1) expression does not sufficiently predict ICI efficacy in NSCLC patients. In recent studies, the tumor immune microenvironment (TIME) was shown to have a central role in lung cancer progression and to affect clinical outcome of patients diagnosed with lung cancer. As development of new therapeutic targets to overcome ICI resistance is a priority, understanding the TIME is important. Recently, a series of studies was conducted to target each component of TIME to improve efficacy of cancer treatment. In this review, important features related to TIME, its heterogeneity and current trends in treatment targeting the component of TIME are discussed. Methods PubMed and PMC were searched from January 1st, 2012 to August 16th, 2022 using the following key words: "NSCLC", "Tumor microenvironment", "Immune", "Metastasis" and "Heterogeneity". Key Content and Findings Heterogeneity in the TIME can be either spatial or temporal. Subsequent to heterogeneous changes in the TIME, treatment of lung cancer can be more challenging because drug resistance is more likely to occur. In terms of the TIME, the main concept for increasing the chance of successful NSCLC treatment is to activate immune responses against tumor cells and inhibit immunosuppressive activities. In addition, relevant research is focused on normalizing an otherwise aberrant TIME in NSCLC patients. Potential therapeutic targets include immune cells, cytokine interactions, and non-immune cells such as fibroblasts or vessels. Conclusions In management of lung cancer, understanding TIME and its heterogeneity is significant to treatment outcomes. Ongoing trials including various treatment modalities such as radiotherapy, cytotoxic chemotherapy, and anti-angiogenic treatment and regimens inhibiting other immunoinhibitory molecules are promising.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eunyoung Lee
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Yun Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, Republic of Korea
| | - Hyeong Jun Cho
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Hyuck Ahn
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yongki Hwang
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
22
|
Boosting In-Vivo Anti-Tumor Immunity with an Oral Microparticulate Breast Cancer Vaccine and Low-Dose Cyclophosphamide. Vaccines (Basel) 2023; 11:vaccines11030543. [PMID: 36992127 DOI: 10.3390/vaccines11030543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Tumor cells express antigens that should induce immune-mediated rejection; however, spontaneous rejection of established tumors is rare. Recent evidence suggests that patients suffering from cancer exhibit an elevation in regulatory T cells population, a subset of CD4+ T cells, which suppress tumor recognition and elimination by cytotoxic T cells. This study investigates immunotherapeutic strategies to overcome the immunosuppressive effects exerted by regulatory T cells. A novel immunotherapeutic strategy was developed by simultaneous administration of oral microparticulate breast cancer vaccines and cyclophosphamide, a regulatory T cell inhibitor. Breast cancer vaccine microparticles were prepared by spray drying, and administered orally to female mice inoculated with 4TO7 murine breast cancer cells in combination with a low dose of intraperitoneally administered cyclophosphamide. Mice receiving the combination of vaccine microparticles and cyclophosphamide exhibited maximal tumor regression and the highest survival rate compared with the control groups. This study highlights the importance of cancer vaccination along with regulatory T cell depletion in cancer therapy, and suggests that a low dose of cyclophosphamide that specifically and significantly depletes regulatory T cells may be a highly effective immunotherapeutic strategy for the treatment of cancer.
Collapse
|
23
|
TNFR2 antagonistic antibody induces the death of tumor infiltrating CD4 +Foxp3 + regulatory T cells. Cell Oncol (Dordr) 2023; 46:167-177. [PMID: 36369606 DOI: 10.1007/s13402-022-00742-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND TNFR2 expression is a characteristic of highly potent immunosuppressive tumor infiltrating CD4+Foxp3+ regulatory T cells (Tregs). There is compelling evidence that TNF through TNFR2 preferentially stimulates the activation and expansion of Tregs. We and others, therefore, proposed that targeting TNFR2 may provide a novel strategy in cancer immunotherapy. Several studies have shown the effect of TNFR2 antagonistic antibodies in different tumor models. However, the exact action of the TNFR2 antibody on Tregs remained understood. METHOD TY101, an anti-murine TNFR2 antibody, was used to examine the effect of TNFR2 blockade on Treg proliferation and viability in vitro. The role of TNFR2 on Treg viability was further validated by TNFR2 knockout mice and in the TY101 antagonistic antibody-treated mouse tumor model. RESULTS In this study, we found that an anti-mouse TNFR2 antibody TY101 could inhibit TNF-induced proliferative expansion of Tregs, indicative of an antagonistic property. To examine the effect of TY101 antagonistic antibody on Treg viability, we treated unfractionated lymph node (L.N.) cells with Dexamethasone (Dex) which was known to induce T cell death. The result showed that TY101 antagonistic antibody treatment further promoted Treg death in the presence of Dex. This led us to find that TNFR2 expression was crucial for the survival of Tregs. In the mouse EG7 lymphoma model, treatment with TY101 antagonistic antibody potently inhibited tumor growth, resulting in complete regression of the tumor in 60% of mice. The treatment with TY101 antagonistic antibody elicited potent antitumor immune responses in this model, accompanied by enhanced death of Tregs. CONCLUSION This study, therefore, provides clear experimental evidence that TNFR2 antagonistic antibody, TY101, can promote the death of Tregs, and this effect may be attributable to the antitumor effect of TNFR2 antagonistic antibody.
Collapse
|
24
|
Clinical Effects of Immuno-Oncology Therapy on Glioblastoma Patients: A Systematic Review. Brain Sci 2023; 13:brainsci13020159. [PMID: 36831702 PMCID: PMC9953849 DOI: 10.3390/brainsci13020159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
The most prevalent and deadly primary malignant glioma in adults is glioblastoma (GBM), which has a median survival time of about 15 months. Despite the standard of care for glioblastoma, which includes gross total resection, high-dose radiation, and temozolomide chemotherapy, this tumor is still one of the most aggressive and difficult to treat. So, it is critical to find more potent therapies that can help glioblastoma patients have better clinical outcomes. Additionally, the prognosis for recurring malignant gliomas is poor, necessitating the need for innovative therapeutics. Immunotherapy is a rather new treatment for glioblastoma and its effects are not well studied when it is combined with standard chemoradiation therapy. We conducted this study to evaluate different glioblastoma immunotherapy approaches in terms of feasibility, efficacy, and safety. We conducted a computer-assisted literature search of electronic databases for essays that are unique, involve either prospective or retrospective research, and are entirely written and published in English. We examined both observational data and randomized clinical trials. Eighteen studies met the criteria for inclusion. In conclusion, combining immunotherapy with radiochemotherapy and tumor removal is generally possible and safe, and rather effective in the prolongation of survival measures.
Collapse
|
25
|
Devi-Marulkar P, Fastenackels S, Karapentiantz P, Goc J, Germain C, Kaplon H, Knockaert S, Olive D, Panouillot M, Validire P, Damotte D, Alifano M, Murris J, Katsahian S, Lawand M, Dieu-Nosjean MC. Regulatory T cells infiltrate the tumor-induced tertiary lymphoïd structures and are associated with poor clinical outcome in NSCLC. Commun Biol 2022; 5:1416. [PMID: 36566320 PMCID: PMC9789959 DOI: 10.1038/s42003-022-04356-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
On one hand, regulatory T cells (Tregs) play an immunosuppressive activity in most solid tumors but not all. On the other hand, the organization of tumor-infiltrating immune cells into tertiary lymphoid structures (TLS) is associated with long-term survival in most cancers. Here, we investigated the role of Tregs in the context of Non-Small Cell Lung Cancer (NSCLC)-associated TLS. We observed that Tregs show a similar immune profile in TLS and non-TLS areas. Autologous tumor-infiltrating Tregs inhibit the proliferation and cytokine secretion of CD4+ conventional T cells, a capacity which is recovered by antibodies against Cytotoxic T-Lymphocyte-Associated protein-4 (CTLA-4) and Glucocorticoid-Induced TNFR-Related protein (GITR) but not against other immune checkpoint (ICP) molecules. Tregs in the whole tumor, including in TLS, are associated with a poor outcome of NSCLC patients, and combination with TLS-dendritic cells (DCs) and CD8+ T cells allows higher overall survival discrimination. Thus, Targeting Tregs especially in TLS may represent a major challenge in order to boost anti-tumor immune responses initiated in TLS.
Collapse
Affiliation(s)
- Priyanka Devi-Marulkar
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.418596.70000 0004 0639 6384Present Address: Institut Curie, Paris, France
| | - Solène Fastenackels
- grid.462844.80000 0001 2308 1657UMRS1135 Sorbonne Université, Faculté de Médecine Sorbonne Université, Paris, France ,grid.7429.80000000121866389INSERM U1135, Paris, France ,grid.463810.8Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Pierre Karapentiantz
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,Present Address: Inserm, Sorbonne Université, université Paris 13, Laboratoire d’informatique médicale et d’ingénierie des connaissances en e-santé, LIMICS, F-75006 Paris, France
| | - Jérémy Goc
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.5386.8000000041936877XPresent Address: Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology and Immunology and The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, USA
| | - Claire Germain
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,Present Address: Biomunex Pharmaceuticals, Paris, France
| | - Hélène Kaplon
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.418301.f0000 0001 2163 3905Present Address: Translational Medicine Department, Institut de Recherches Internationales Servier, Suresnes, France
| | - Samantha Knockaert
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.418301.f0000 0001 2163 3905Present Address: Translational Medicine Department, Institut de Recherches Internationales Servier, Suresnes, France
| | - Daniel Olive
- Inserm U1068, CNRS, UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France ,grid.463833.90000 0004 0572 0656Laboratory « Immunity and Cancer », Centre de Recherche en Cancérologie de Marseille (CRCM), Marseille, France
| | - Marylou Panouillot
- grid.462844.80000 0001 2308 1657UMRS1135 Sorbonne Université, Faculté de Médecine Sorbonne Université, Paris, France ,grid.7429.80000000121866389INSERM U1135, Paris, France ,grid.463810.8Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Pierre Validire
- grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.418120.e0000 0001 0626 5681Department of Pathology, Institut Mutualiste Montsouris, Paris, France
| | - Diane Damotte
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.411784.f0000 0001 0274 3893Department of Pathology, Assistance Publique-Hôpitaux de Paris (AP-HP), Cochin hospital, Paris, France
| | - Marco Alifano
- grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.50550.350000 0001 2175 4109Department of Thoracic Surgery, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Juliette Murris
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.5328.c0000 0001 2186 3954HeKA, INRIA, Paris, France ,Hôpital Européen Georges-Pompidou, Unité d’Epidémiologie et de Recherche Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Inserm, Centre d’Investigation Clinique 1418, Module Epidémiologie Clinique, Paris, France
| | - Sandrine Katsahian
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.5328.c0000 0001 2186 3954HeKA, INRIA, Paris, France ,Hôpital Européen Georges-Pompidou, Unité d’Epidémiologie et de Recherche Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Inserm, Centre d’Investigation Clinique 1418, Module Epidémiologie Clinique, Paris, France
| | - Myriam Lawand
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- grid.503414.7Sorbonne Université, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.410511.00000 0001 2149 7878Université de Paris, UMRS 1138, Cordeliers Research Center, Paris, France ,grid.417925.cLaboratory “Cancer, Immune Control, and Escape”, Inserm U1138, Cordeliers Research Center, Paris, France ,grid.462844.80000 0001 2308 1657UMRS1135 Sorbonne Université, Faculté de Médecine Sorbonne Université, Paris, France ,grid.7429.80000000121866389INSERM U1135, Paris, France ,grid.463810.8Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| |
Collapse
|
26
|
Gertych A, Walts AE, Cheng K, Liu M, John J, Lester J, Karlan BY, Orsulic S. Dynamic Changes in the Extracellular Matrix in Primary, Metastatic, and Recurrent Ovarian Cancers. Cells 2022; 11:3769. [PMID: 36497028 PMCID: PMC9736731 DOI: 10.3390/cells11233769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) and their extracellular matrix are active participants in cancer progression. While it is known that functionally different subpopulations of CAFs co-exist in ovarian cancer, it is unclear whether certain CAF subsets are enriched during metastatic progression and/or chemotherapy. Using computational image analyses of patient-matched primary high-grade serous ovarian carcinomas, synchronous pre-chemotherapy metastases, and metachronous post-chemotherapy metastases from 42 patients, we documented the dynamic spatiotemporal changes in the extracellular matrix, fibroblasts, epithelial cells, immune cells, and CAF subsets expressing different extracellular matrix components. Among the different CAF subsets, COL11A1+ CAFs were associated with linearized collagen fibers and exhibited the greatest enrichment in pre- and post-chemotherapy metastases compared to matched primary tumors. Although pre- and post-chemotherapy metastases were associated with increased CD8+ T cell infiltration, the infiltrate was not always evenly distributed between the stroma and cancer cells, leading to an increased frequency of the immune-excluded phenotype where the majority of CD8+ T cells are present in the tumor stroma but absent from the tumor parenchyma. Overall, most of the differences in the tumor microenvironment were observed between primary tumors and metastases, while fewer differences were observed between pre- and post-treatment metastases. These data suggest that the tumor microenvironment is largely determined by the primary vs. metastatic location of the tumor while chemotherapy does not have a significant impact on the host microenvironment.
Collapse
Affiliation(s)
- Arkadiusz Gertych
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Faculty of Biomedical Engineering, Silesian University of Technology, 44-100 Zabrze, Poland
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Keyi Cheng
- Department of Mathematics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Manyun Liu
- Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro, GA 30458, USA
| | - Joshi John
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| | - Jenny Lester
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
27
|
Rašková M, Lacina L, Kejík Z, Venhauerová A, Skaličková M, Kolář M, Jakubek M, Rosel D, Smetana K, Brábek J. The Role of IL-6 in Cancer Cell Invasiveness and Metastasis-Overview and Therapeutic Opportunities. Cells 2022; 11:3698. [PMID: 36429126 PMCID: PMC9688109 DOI: 10.3390/cells11223698] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Interleukin 6 (IL-6) belongs to a broad class of cytokines involved in the regulation of various homeostatic and pathological processes. These activities range from regulating embryonic development, wound healing and ageing, inflammation, and immunity, including COVID-19. In this review, we summarise the role of IL-6 signalling pathways in cancer biology, with particular emphasis on cancer cell invasiveness and metastasis formation. Targeting principal components of IL-6 signalling (e.g., IL-6Rs, gp130, STAT3, NF-κB) is an intensively studied approach in preclinical cancer research. It is of significant translational potential; numerous studies strongly imply the remarkable potential of IL-6 signalling inhibitors, especially in metastasis suppression.
Collapse
Affiliation(s)
- Magdalena Rašková
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Lukáš Lacina
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Zdeněk Kejík
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Anna Venhauerová
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Markéta Skaličková
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Michal Kolář
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, 140 00 Prague, Czech Republic
| | - Milan Jakubek
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Karel Smetana
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| |
Collapse
|
28
|
Berg-Larsen A, Mobergslien A, Moen I, Petros G, Kristian A, Gunvaldsen KS, Cruciani V, Wickstroem K, Bjerke RM, Karlsson J, Cuthbertson A. Tumor growth inhibition and immune system activation following treatment with thorium-227 conjugates and PD-1 check-point inhibition in the MC-38 murine model. Front Med (Lausanne) 2022; 9:1033303. [DOI: 10.3389/fmed.2022.1033303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
Targeted thorium-227 conjugates comprise the combination of a monoclonal antibody with specificity for a tumor cell antigen and a 3,2-HOPO chelator enabling complexation of thorium-227 (Th-227). The radiolabeled conjugate functions as an effective delivery system of alpha-particle radiation to the surface of the tumor cell inducing difficult to repair complex DNA damage and cell death. In addition, the mechanism of action of targeted alpha therapy (TAT) appears to involve a significant component linked to stimulation of the immune system. We report herein evidence of immune activation and long-lasting immune protection of a TAT in a syngeneic model using the MC-38 murine cell line. Firstly, MC-38 cells were irradiated ex vivo with the thorium labeled antibody before subcutaneous implantation into mice. These mice were then rechallenged with MC-38 cells contra-laterally. In the group receiving irradiated cells, 9 out of 10 animals had no measurable tumor growth compared to aggressive tumor growth in the control group. Secondly, in an efficacy study, 500 kBq/kg of thorium labeled antibody alone or in combination with PD-1 checkpoint inhibitor gave statistically significant tumor growth inhibition compared to vehicle control. Animals with no measurable tumors were once again rechallenged contra-laterally with MC-38 cells. The re-growth of tumors was significantly delayed (approx. 60 days) in the treatment group compared to age-matched controls (approx. 30 days) in the monotherapy group. Interestingly, in the TAT/ PD-1 combination group no re-growth was observed demonstrating the potential of combining a TAT with checkpoint inhibition therapy. Finally, tumors were excised from treated mice and analyzed by flow cytometry and immunohistochemistry (IHC). Analysis revealed significant infiltration of CD8+ T-cells and mature dendritic cells compared to vehicle controls. Together these results indicated that an ongoing immune response from treatment with alpha radiation could be enhanced by check-point inhibition.
Collapse
|
29
|
Regulatory T Cells in Ovarian Carcinogenesis and Future Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14225488. [PMID: 36428581 PMCID: PMC9688690 DOI: 10.3390/cancers14225488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022] Open
Abstract
Regulatory T cells (Tregs) have been shown to play a role in the development of solid tumors. A better understanding of the biology of Tregs, immune suppression by Tregs, and how cancer developed with the activity of Tregs has facilitated the development of strategies used to improve immune-based therapy. In ovarian cancer, Tregs have been shown to promote cancer development and resistance at different cancer stages. Understanding the various Treg-mediated immune escape mechanisms provides opportunities to establish specific, efficient, long-lasting anti-tumor immunity. Here, we review the evidence of Treg involvement in various stages of ovarian cancer. We further provide an overview of the current and prospective therapeutic approaches that arise from the modulation of Treg-related tumor immunity at those specific stages. Finally, we propose combination strategies of Treg-related therapies with other anti-tumor therapies to improve clinical efficacy and overcome tumor resistance in ovarian cancer.
Collapse
|
30
|
Laba S, Mallett G, Amarnath S. The depths of PD-1 function within the tumor microenvironment beyond CD8 + T cells. Semin Cancer Biol 2022; 86:1045-1055. [PMID: 34048897 DOI: 10.1016/j.semcancer.2021.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/30/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Programmed cell death-1 (PD-1; CD279) is a cell surface receptor that is expressed in both innate and adaptive immune cells. The role of PD-1 in adaptive immune cells, specifically in CD8+ T cells, has been thoroughly investigated but its significance in other immune cells is yet to be well established. This review will address the role of PD-1 based therapies in enhancing non-CD8+ T cell immune responses within cancer. Specifically, the expression and function of PD-1 in non-CD8+ immune cell compartments such as CD4+ T helper cell subsets, myeloid cells and innate lymphoid cells (ILCs) will be discussed. By understanding the immune cell specific function of PD-1 within tissue resident innate and adaptive immune cells, it will be possible to stratify patients for PD-1 based therapies for both immunogeneic and non-immunogeneic neoplastic disorders. With this knowledge from fundamental and translational studies, PD-1 based therapies can be utilized to enhance T cell independent immune responses in cancers.
Collapse
Affiliation(s)
- Stephanie Laba
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, United Kingdom.
| | - Grace Mallett
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, United Kingdom
| | - Shoba Amarnath
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, United Kingdom.
| |
Collapse
|
31
|
Hudry D, Le Guellec S, Meignan S, Bécourt S, Pasquesoone C, El Hajj H, Martínez-Gómez C, Leblanc É, Narducci F, Ladoire S. Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints. Cancers (Basel) 2022; 14:5332. [PMID: 36358750 PMCID: PMC9656626 DOI: 10.3390/cancers14215332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Epithelial ovarian cancers (EOC) are often diagnosed at an advanced stage with carcinomatosis and a poor prognosis. First-line treatment is based on a chemotherapy regimen combining a platinum-based drug and a taxane-based drug along with surgery. More than half of the patients will have concern about a recurrence. To improve the outcomes, new therapeutics are needed, and diverse strategies, such as immunotherapy, are currently being tested in EOC. To better understand the global immune contexture in EOC, several studies have been performed to decipher the landscape of tumor-infiltrating lymphocytes (TILs). CD8+ TILs are usually considered effective antitumor immune effectors that immune checkpoint inhibitors can potentially activate to reject tumor cells. To synthesize the knowledge of TILs in EOC, we conducted a review of studies published in MEDLINE or EMBASE in the last 10 years according to the PRISMA guidelines. The description and role of TILs in EOC prognosis are reviewed from the published data. The links between TILs, DNA repair deficiency, and ICs have been studied. Finally, this review describes the role of TILs in future immunotherapy for EOC.
Collapse
Affiliation(s)
- Delphine Hudry
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Solenn Le Guellec
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Samuel Meignan
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille University, F-59000 Lille, France
| | - Stéphanie Bécourt
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Camille Pasquesoone
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Houssein El Hajj
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | | | - Éric Leblanc
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Fabrice Narducci
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, F-21000 Dijon, France
- INSERM, CRI-866 Faculty of Medicine, F-21000 Dijon, France
| |
Collapse
|
32
|
Immune Tumor Microenvironment in Ovarian Cancer Ascites. Int J Mol Sci 2022; 23:ijms231810692. [PMID: 36142615 PMCID: PMC9504085 DOI: 10.3390/ijms231810692] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer (OC) has a specific type of metastasis, via transcoelomic, and most of the patients are diagnosed at advanced stages with multiple tumors spread within the peritoneal cavity. The role of Malignant Ascites (MA) is to serve as a transporter of tumor cells from the primary location to the peritoneal wall or to the surface of the peritoneal organs. MA comprise cellular components with tumor and non-tumor cells and acellular components, creating a unique microenvironment capable of modifying the tumor behavior. These microenvironment factors influence tumor cell proliferation, progression, chemoresistance, and immune evasion, suggesting that MA play an active role in OC progression. Tumor cells induce a complex immune suppression that neutralizes antitumor immunity, leading to disease progression and treatment failure, provoking a tumor-promoting environment. In this review, we will focus on the High-Grade Serous Carcinoma (HGSC) microenvironment with special attention to the tumor microenvironment immunology.
Collapse
|
33
|
Bound NT, Vandenberg CJ, Kartikasari AER, Plebanski M, Scott CL. Improving PARP inhibitor efficacy in high-grade serous ovarian carcinoma: A focus on the immune system. Front Genet 2022; 13:886170. [PMID: 36159999 PMCID: PMC9505691 DOI: 10.3389/fgene.2022.886170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/05/2022] [Indexed: 12/03/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a genomically unstable malignancy responsible for over 70% of all deaths due to ovarian cancer. With roughly 50% of all HGSOC harboring defects in the homologous recombination (HR) DNA repair pathway (e.g., BRCA1/2 mutations), the introduction of poly ADP-ribose polymerase inhibitors (PARPi) has dramatically improved outcomes for women with HR defective HGSOC. By blocking the repair of single-stranded DNA damage in cancer cells already lacking high-fidelity HR pathways, PARPi causes the accumulation of double-stranded DNA breaks, leading to cell death. Thus, this synthetic lethality results in PARPi selectively targeting cancer cells, resulting in impressive efficacy. Despite this, resistance to PARPi commonly develops through diverse mechanisms, such as the acquisition of secondary BRCA1/2 mutations. Perhaps less well documented is that PARPi can impact both the tumour microenvironment and the immune response, through upregulation of the stimulator of interferon genes (STING) pathway, upregulation of immune checkpoints such as PD-L1, and by stimulating the production of pro-inflammatory cytokines. Whilst targeted immunotherapies have not yet found their place in the clinic for HGSOC, the evidence above, as well as ongoing studies exploring the synergistic effects of PARPi with immune agents, including immune checkpoint inhibitors, suggests potential for targeting the immune response in HGSOC. Additionally, combining PARPi with epigenetic-modulating drugs may improve PARPi efficacy, by inducing a BRCA-defective phenotype to sensitise resistant cancer cells to PARPi. Finally, invigorating an immune response during PARPi therapy may engage anti-cancer immune responses that potentiate efficacy and mitigate the development of PARPi resistance. Here, we will review the emerging PARPi literature with a focus on PARPi effects on the immune response in HGSOC, as well as the potential of epigenetic combination therapies. We highlight the potential of transforming HGSOC from a lethal to a chronic disease and increasing the likelihood of cure.
Collapse
Affiliation(s)
- Nirashaa T. Bound
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Cassandra J. Vandenberg
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Apriliana E. R. Kartikasari
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Magdalena Plebanski
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Clare L. Scott
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Royal Women’s Hospital, Parkville, VIC, Australia
| |
Collapse
|
34
|
OATD-02 Validates the Benefits of Pharmacological Inhibition of Arginase 1 and 2 in Cancer. Cancers (Basel) 2022; 14:cancers14163967. [PMID: 36010962 PMCID: PMC9406419 DOI: 10.3390/cancers14163967] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Arginase 1 and 2 are drivers of multiple immunosuppressive mechanisms and tumour-specific metabolic adaptations. Pharmacological inhibition of extracellular ARG1 has shown antitumour efficacy in various syngeneic tumour models, however, the importance of ARG2 as a therapeutic target has only been demonstrated by genetic deletion studies. This is the first study validating the benefits of pharmacological inhibition of ARG2 in cancer. Our work describes OATD-02 as a potent dual ARG1/ARG2 inhibitor with a cellular activity (necessary for targeting ARG2) exhibiting immunomodulatory and direct antitumour efficacy in animal models. Our results present OATD-02 as an attractive option for combination with other immunotherapeutics, such as PD-1/PD-L1 antibodies or IDO1 inhibitors, especially in the therapy of particularly resistant hypoxic tumours. The presented findings provided the rationale for planning first-in-human clinical trials for OATD-02 in cancer patients. Abstract Background: Arginases play essential roles in metabolic pathways, determining the fitness of both immune and tumour cells. Along with the previously validated role of ARG1 in cancer, the particular significance of ARG2 as a therapeutic target has emerged as its levels correlate with malignant phenotype and poor prognosis. These observations unveil arginases, and specifically ARG2, as well-validated and promising therapeutic targets. OATD-02, a new boronic acid derivative, is the only dual inhibitor, which can address the benefits of pharmacological inhibition of arginase 1 and 2 in cancer. Methods: The inhibitory activity of OATD-02 was determined using recombinant ARG1 and ARG2, as well as in a cellular system using primary hepatocytes and macrophages. In vivo antitumor activity was determined in syngeneic models of colorectal and kidney carcinomas (CT26 and Renca, respectively), as well as in an ARG2-dependent xenograft model of leukaemia (K562). Results: OATD-02 was shown to be a potent dual (ARG1/ARG2) arginase inhibitor with a cellular activity necessary for targeting ARG2. Compared to a reference inhibitor with predominant extracellular activity towards ARG1, we have shown improved and statistically significant antitumor efficacy in the CT26 model and an immunomodulatory effect reflected by Treg inhibition in the Renca model. Importantly, OATD-02 had a superior activity when combined with other immunotherapeutics. Finally, OATD-02 effectively inhibited the proliferation of human K562 leukemic cells both in vitro and in vivo. Conclusions: OATD-02 is a potent small-molecule arginase inhibitor with optimal drug-like properties, including PK/PD profile. Excellent activity against intracellular ARG2 significantly distinguishes OATD-02 from other arginase inhibitors. OATD-02 represents a very promising drug candidate for the combined treatment of tumours, and is the only pharmacological tool that can effectively address the benefits of ARG1/ARG2 inhibition. OATD-02 will enter clinical trials in cancer patients in 2022.
Collapse
|
35
|
Li H, Zheng X, Gao J, Leung KS, Wong MH, Yang S, Liu Y, Dong M, Bai H, Ye X, Cheng L. Whole transcriptome analysis reveals non-coding RNA's competing endogenous gene pairs as novel form of motifs in serous ovarian cancer. Comput Biol Med 2022; 148:105881. [DOI: 10.1016/j.compbiomed.2022.105881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/30/2022] [Accepted: 07/16/2022] [Indexed: 11/03/2022]
|
36
|
Combinatorial immunotherapies overcome MYC-driven immune evasion in triple negative breast cancer. Nat Commun 2022; 13:3671. [PMID: 35760778 PMCID: PMC9237085 DOI: 10.1038/s41467-022-31238-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/09/2022] [Indexed: 12/14/2022] Open
Abstract
Few patients with triple negative breast cancer (TNBC) benefit from immune checkpoint inhibitors with complete and durable remissions being quite rare. Oncogenes can regulate tumor immune infiltration, however whether oncogenes dictate diminished response to immunotherapy and whether these effects are reversible remains poorly understood. Here, we report that TNBCs with elevated MYC expression are resistant to immune checkpoint inhibitor therapy. Using mouse models and patient data, we show that MYC signaling is associated with low tumor cell PD-L1, low overall immune cell infiltration, and low tumor cell MHC-I expression. Restoring interferon signaling in the tumor increases MHC-I expression. By combining a TLR9 agonist and an agonistic antibody against OX40 with anti-PD-L1, mice experience tumor regression and are protected from new TNBC tumor outgrowth. Our findings demonstrate that MYC-dependent immune evasion is reversible and druggable, and when strategically targeted, may improve outcomes for patients treated with immune checkpoint inhibitors.
Collapse
|
37
|
Rodriguez GM, Galpin KJ, Cook DP, Yakubovich E, Maranda V, Macdonald EA, Wilson-Sanchez J, Thomas AL, Burdette JE, Vanderhyden BC. The Tumor Immune Profile of Murine Ovarian Cancer Models: An Essential Tool For Ovarian Cancer Immunotherapy Research. CANCER RESEARCH COMMUNICATIONS 2022; 2:417-433. [PMID: 36311166 PMCID: PMC9616009 DOI: 10.1158/2767-9764.crc-22-0017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/07/2022] [Accepted: 05/18/2022] [Indexed: 06/16/2023]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic cancer with an imperative need for new treatments. Immunotherapy has had marked success in some cancer types; however, clinical trials studying the efficacy of immune checkpoint inhibitors for the treatment of EOC benefited less than 15% of patients. Given that EOC develops from multiple tissues in the reproductive system and metastasizes widely throughout the peritoneal cavity, responses to immunotherapy are likely hindered by heterogeneous tumor microenvironments (TME) containing a variety of immune profiles. To fully characterize and compare syngeneic model systems that may reflect this diversity, we determined the immunogenicity of six ovarian tumor models in vivo, the T and myeloid profile of orthotopic tumors and the immune composition and cytokine profile of ascites, by single-cell RNA sequencing, flow cytometry and IHC. The selected models reflect the different cellular origins of EOC (ovarian and fallopian tube epithelium) and harbor mutations relevant to human disease, including Tp53 mutation, PTEN suppression, and constitutive KRAS activation. ID8-p53-/- and ID8-C3 tumors were most highly infiltrated by T cells, whereas STOSE and MOE-PTEN/KRAS tumors were primarily infiltrated by tumor associated macrophages and were unique in MHC class I and II expression. MOE-PTEN/KRAS tumors were capable of forming T cell clusters. This panel of well-defined murine EOC models reflects some of the heterogeneity found in human disease and can serve as a valuable resource for studies that aim to test immunotherapies, explore the mechanisms of immune response to therapy, and guide selection of treatments for patient populations.
Collapse
Affiliation(s)
- Galaxia M. Rodriguez
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Kristianne J.C. Galpin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David P. Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Edward Yakubovich
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Vincent Maranda
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Elizabeth A. Macdonald
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Juliette Wilson-Sanchez
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Anjali L. Thomas
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Barbara C. Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
38
|
Elkoshi Z. On the Prognostic Power of Tumor-Infiltrating Lymphocytes – A Critical Commentary. Front Immunol 2022; 13:892543. [PMID: 35634289 PMCID: PMC9133417 DOI: 10.3389/fimmu.2022.892543] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor-infiltrating lymphocytes are extensively used as prognostic biomarkers in cancer. Regulatory T cells (Tregs) or CD8+ T cells frequencies in tumor site, or their ratio, are the most common markers used to assess prognosis. This work offers a possible explanation for the opposite correlations between intra-tumoral Tregs and survival, associated with different types of cancer. The complexity involved with the selection of a preferred marker, including the effect of variability, is presented and discussed. The lymphocytes frequency ratio is proposed as the marker of choice in most types of cancer. The ratio correlates directly with survival, irrespective of cancer type and is also less variable than the frequencies of each of the two lymphocytes, if these frequencies correlate with each other in the tumor microenvironment. However, if the frequency of one of the two lymphocytes is highly variable, abandoning the ratio in favor of the lymphocyte with less variable frequency will improve correlation with survival, especially when the intra-tumoral frequencies of the two species are inversely correlated. It is plausible, that the best prognostic marker selected this way, will be also be the best predictor of checkpoint inhibitor therapy success.
Collapse
|
39
|
Jiang X, Wang J, Zheng X, Liu Z, Zhang X, Li Y, Wilhelm J, Cao J, Huang G, Zhang J, Sumer B, Lea J, Lu Z, Gao J, Luo M. Intratumoral administration of STING-activating nanovaccine enhances T cell immunotherapy. J Immunother Cancer 2022; 10:jitc-2021-003960. [PMID: 35623658 PMCID: PMC9150169 DOI: 10.1136/jitc-2021-003960] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cancer vaccines are able to achieve tumor-specific immune editing in early-phase clinical trials. However, the infiltration of cytotoxic T cells into immune-deserted tumors is still a major limiting factor. An optimized vaccine approach to induce antigen-specific T cells that can perform robust tumor infiltration is important to accelerate their clinical translation. We previously developed a STING-activating PC7A nanovaccine that produces a strong anti-tumor T cell response on subcutaneous injection. This study systematically investigated the impact of administration methods on the performance of nanovaccines. METHODS Tumor growth inhibition by intratumoral delivery and subcutaneous delivery of nanovaccine was investigated in TC-1 human papillomavirus-induced cancer model and B16-OVA melanoma model. Nanovaccine distribution in vivo was detected by clinical camera imaging, systemic T cell activation and tumor infiltration were tested by in vivo cytotoxicity killing assay and flow cytometry. For mechanism analysis, T cell recruitment was investigated by in vivo migration blocking assay, multiplex chemokine array, flow cytometry, RT-qPCR, chemotaxis assay and gene knockout mice. RESULTS Nanovaccine administration was found to alter T cell production and infiltration in tumors. Intratumoral delivery of nanovaccines displayed superior antitumor effects in multiple tumor models compared with subcutaneous delivery. Mechanistic investigation revealed that intratumoral administration of the nanovaccine significantly increased the infiltration of antigen-specific T cells in TC-1 tumors, despite the lower systemic levels of T cells compared with subcutaneous injection. The inhibition of tumor growth by nanovaccines is primarily dependent on CD8+ cytotoxic T cells. Nanovaccine accumulation in tumors upregulates CXCL9 expression in myeloid cells in a STING dependent manner, leading to increased recruitment of IFNγ-expressing CD8+ T cells from the periphery, and IFNγ reciprocally stimulates CXCL9 expression in myeloid cells, resulting in positive feedback between myeloid-CXCL9 and T cell-IFNγ to promote T cell recruitment. However, the STING agonist alone could not sustain this effect in the presence of a systemic deficiency in antigen-specific T cells. CONCLUSIONS Our results demonstrate that intratumoral administration of PC7A nanovaccine achieved stronger antitumor immunity and efficacy over subcutaneous injection. These data suggest intratumoral administration should be included in the therapeutic design in the clinical use of nanovaccine.
Collapse
Affiliation(s)
- Xiaoyi Jiang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Pharmacology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xichen Zheng
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhida Liu
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi, China
| | - Xinyu Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yuwei Li
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jonathan Wilhelm
- Department of Pharmacology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jun Cao
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Gang Huang
- Department of Pharmacology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jinlan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Baran Sumer
- Department of Otolaryngology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jayanthi Lea
- Department of Obstetrics and Gynecology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zhigang Lu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China .,The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China.,Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinming Gao
- Department of Pharmacology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA .,Department of Otolaryngology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Min Luo
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Revenko A, Carnevalli LS, Sinclair C, Johnson B, Peter A, Taylor M, Hettrick L, Chapman M, Klein S, Solanki A, Gattis D, Watt A, Hughes AM, Magiera L, Kar G, Ireland L, Mele DA, Sah V, Singh M, Walton J, Mairesse M, King M, Edbrooke M, Lyne P, Barry ST, Fawell S, Goldberg FW, MacLeod AR. Direct targeting of FOXP3 in Tregs with AZD8701, a novel antisense oligonucleotide to relieve immunosuppression in cancer. J Immunother Cancer 2022; 10:jitc-2021-003892. [PMID: 35387780 PMCID: PMC8987763 DOI: 10.1136/jitc-2021-003892] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The Regulatory T cell (Treg) lineage is defined by the transcription factor FOXP3, which controls immune-suppressive gene expression profiles. Tregs are often recruited in high frequencies to the tumor microenvironment where they can suppress antitumor immunity. We hypothesized that pharmacological inhibition of FOXP3 by systemically delivered, unformulated constrained ethyl-modified antisense oligonucleotides could modulate the activity of Tregs and augment antitumor immunity providing therapeutic benefit in cancer models and potentially in man. METHODS We have identified murine Foxp3 antisense oligonucleotides (ASOs) and clinical candidate human FOXP3 ASO AZD8701. Pharmacology and biological effects of FOXP3 inhibitors on Treg function and antitumor immunity were tested in cultured Tregs and mouse syngeneic tumor models. Experiments were controlled by vehicle and non-targeting control ASO groups as well as by use of multiple independent FOXP3 ASOs. Statistical significance of biological effects was evaluated by one or two-way analysis of variance with multiple comparisons. RESULTS AZD8701 demonstrated a dose-dependent knockdown of FOXP3 in primary Tregs, reduction of suppressive function and efficient target downregulation in humanized mice at clinically relevant doses. Surrogate murine FOXP3 ASO, which efficiently downregulated Foxp3 messenger RNA and protein levels in primary Tregs, reduced Treg suppressive function in immune suppression assays in vitro. FOXP3 ASO promoted more than 70% reduction in FOXP3 levels in Tregs in vitro and in vivo, strongly modulated Treg effector molecules (eg, ICOS, CTLA-4, CD25 and 4-1BB), and augmented CD8+ T cell activation and produced antitumor activity in syngeneic tumor models. The combination of FOXP3 ASOs with immune checkpoint blockade further enhanced antitumor efficacy. CONCLUSIONS Antisense inhibitors of FOXP3 offer a promising novel cancer immunotherapy approach. AZD8701 is being developed clinically as a first-in-class FOXP3 inhibitor for the treatment of cancer currently in Ph1a/b clinical trial (NCT04504669).
Collapse
Affiliation(s)
| | | | | | - Ben Johnson
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | | | | | - Melissa Chapman
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | | | | | - Andrew Watt
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | | | - Gozde Kar
- Oncology R&D, AstraZeneca, Cambridge, UK
| | | | | | - Vasu Sah
- Oncology R&D, AstraZeneca, Waltham, MA, USA
| | | | | | | | | | | | - Paul Lyne
- Oncology R&D, AstraZeneca, Waltham, MA, USA
| | | | | | | | | |
Collapse
|
41
|
Moatti A, Debesset A, Pilon C, Beldi-Ferchiou A, Leclerc M, Redjoul R, Charlotte F, To NH, Bak A, Belkacemi Y, Salomon BL, Issa F, Michonneau D, Maury S, Cohen JL, Thiolat A. TNFR2 blockade of regulatory T cells unleashes an antitumor immune response after hematopoietic stem-cell transplantation. J Immunother Cancer 2022; 10:jitc-2021-003508. [PMID: 35387779 PMCID: PMC8987798 DOI: 10.1136/jitc-2021-003508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 12/13/2022] Open
Abstract
Background Targeting immune checkpoints that inhibit antitumor immune responses has emerged as a powerful new approach to treat cancer. We recently showed that blocking the tumor necrosis factor receptor-type 2 (TNFR2) pathway induces the complete loss of the protective function of regulatory T cells (Tregs) in a model of graft-versus-host disease (GVHD) prevention that relies on Treg-based cell therapy. Here, we tested the possibility of amplifying the antitumor response by targeting TNFR2 in a model of tumor relapse following hematopoietic stem-cell transplantation, a clinical situation for which the need for efficient therapeutic options is still unmet. Method We developed appropriate experimental conditions that mimic patients that relapsed from their initial hematological malignancy after hematopoietic stem-cell transplantation. This consisted of defining in allogeneic bone marrow transplantation models developed in mice, the maximum number of required tumor cells and T cells to infuse into recipient mice to develop a model of tumor relapse without inducing GVHD. We next evaluated whether anti-TNFR2 treatment could trigger alloreactivity and consequently antitumor immune response. In parallel, we also studied the differential expression of TNFR2 on T cells including Treg from patients in post-transplant leukemia relapse and in patients developing GVHD. Results Using experimental conditions in which neither donor T cells nor TNFR2-blocking antibody per se have any effect on tumor relapse, we observed that the coadministration of a suboptimal number of T cells and an anti-TNFR2 treatment can trigger alloreactivity and subsequently induce a significant antitumor effect. This was associated with a reduced percentage of activated CD4+ and CD8+ Tregs. Importantly, human Tregs over-expressed TNFR2 relative to conventional T cells in healthy donors and in patients experiencing leukemia relapse or cortico-resistant GVHD after hematopoietic stem cell transplantation. Conclusions These results highlight TNFR2 as a new target molecule for the development of immunotherapies to treat blood malignancy relapse, used either directly in grafted patients or to enhance donor lymphocyte infusion strategies. More widely, they open the door for new perspectives to amplify antitumor responses against solid cancers by directly targeting Tregs through their TNFR2 expression.
Collapse
Affiliation(s)
- Audrey Moatti
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France.,CIC Biotherapy, GHU Chenevier Mondor, Créteil, France
| | - Anais Debesset
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France
| | | | | | - Mathieu Leclerc
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France.,Service d'hématologie Clinique, GHU Chenevier Mondor, Créteil, France
| | - Rabah Redjoul
- Service d'hématologie Clinique, GHU Chenevier Mondor, Créteil, France
| | - Frederic Charlotte
- Service d'anatomopathologie, University Hospital Pitié Salpêtrière, Paris, France
| | - Nhu Hanh To
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France.,Service d'oncologie-radiothérapie, GHU Chenevier Mondor, Créteil, France
| | - Adeline Bak
- Service d'oncologie-radiothérapie, GHU Chenevier Mondor, Créteil, France
| | - Yazid Belkacemi
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France.,Service d'oncologie-radiothérapie, GHU Chenevier Mondor, Créteil, France
| | - Benoît Laurent Salomon
- INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Paris, France
| | - Fadi Issa
- Transplantation Research Immunology Group, University of Oxford Nuffield Department of Surgical Sciences, Oxford, UK
| | | | - Sebastien Maury
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France.,Service d'hématologie Clinique, GHU Chenevier Mondor, Créteil, France
| | - José Laurent Cohen
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France .,CIC Biotherapy, GHU Chenevier Mondor, Créteil, France
| | - Allan Thiolat
- INSERM, IMRB, Université Paris-Est Créteil Val de Marne, Créteil, France
| |
Collapse
|
42
|
Yu B, Yoo D, Kim KH, Kim TW, Park S, Kim Y, Son Y, Kim J, Noh I, Whang C, Chung J, Jon S. Effective Combination Immunotherapy through Vessel Normalization Using a Cancer‐Targeting Antiangiogenic Peptide–Antibody Hybrid. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Byeongjun Yu
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Dohyun Yoo
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Ki Hyun Kim
- Department of Biochemistry and Molecular Biology Seoul National University College of Medicine 103 Daehak‐ro Seoul 03080 Republic of Korea
| | - Tae Woo Kim
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Seho Park
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Yujin Kim
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Youngju Son
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Jinjoo Kim
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Ilkoo Noh
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Chang‐Hee Whang
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology Seoul National University College of Medicine 103 Daehak‐ro Seoul 03080 Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences KAIST Institute for the BioCentury Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
- Center for Precision Bio‐Nanomedicine Korea Advanced Institute of Sciences and Technology (KAIST) 291 Daehak‐ro Daejeon 34141 Korea
| |
Collapse
|
43
|
Orr B, Mahdi H, Fang Y, Strange M, Uygun I, Rana M, Zhang L, Suarez Mora A, Pusateri A, Elishaev E, Kang C, Tseng G, Gooding W, Edwards RP, Kalinski P, Vlad AM. Phase I trial combining chemokine-targeting with loco-regional chemo-immunotherapy for recurrent, platinum-sensitive ovarian cancer shows induction of CXCR3 ligands and markers of type 1 immunity. Clin Cancer Res 2022; 28:2038-2049. [PMID: 35046055 DOI: 10.1158/1078-0432.ccr-21-3659] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/03/2021] [Accepted: 01/12/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Increased prevalence of cytotoxic T lymphocytes (CTL) in the tumor microenvironment (TME) predicts positive outcomes in patients with epithelial ovarian cancer (EOC), while the regulatory Treg cells predict poor outcomes. Guided by the synergistic activity of TLR3 ligands, interferon-a (IFNa) and cyclooxygenase-2 (COX-2) blockers in selectively enhancing CTL-attractants but suppressing Treg-attractants, we tested a novel intraperitoneal (IP) chemo-immunotherapy combination, to assess its tolerability and TME-modulatory impact in patients with recurrent EOC. METHODS Twelve patients were enrolled in phase I portion of the trial NCT02432378, and treated with IP cisplatin, IP rintatolimod (dsRNA, TLR3 ligand) and oral celecoxib (COX-2 blocker). Patients in cohorts 2, 3 and 4 also received IP IFNa at 2, 6 and 18 million units (MU), respectively. Primary objectives were to evaluate safety, identify phase 2 recommended dose (P2RD) and characterize changes in the immune TME. Peritoneal resident cells and IP wash fluid were profiled via NanoString and Meso Scale Discovery (MSD) multiplex assay, respectively. RESULTS The P2RD of IFNa was 6 MU. Median progression-free and overall survival were 8.4 and 30 months, respectively. Longitudinal sampling of the peritoneal cavity via IP washes demonstrated local upregulation of interferon-stimulated genes (ISG), including CTL-attracting chemokines (CXCL-9, -10, -11), MHC I/II, perforin and granzymes. These changes were present two days post chemokine modulation and subsided within one week. CONCLUSION The chemokine-modulating IP-CITC is safe, tolerable, and associated with ISG changes that favor CTL chemoattraction and function. This combination (plus DC vaccine) will be tested in a phase II trial.
Collapse
Affiliation(s)
- Brian Orr
- Gynecologic Oncology, Medical University of South Carolina
| | - Haider Mahdi
- Gynecologic Oncology, University of Pittsburgh Medical Center
| | - Yusi Fang
- Biostatistics, University of Pittsburgh, Graduate School of Public Health
| | | | - Ibrahim Uygun
- Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute
| | - Mainpal Rana
- Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine
| | - Lixin Zhang
- Immunology, University of Pittsburgh School of Medicine
| | | | | | - Esther Elishaev
- Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh
| | - Chaeryon Kang
- Biostatistics, University of Pittsburgh Graduate School of Public Health
| | | | | | - Robert P Edwards
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh
| | | | - Anda M Vlad
- Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine
| |
Collapse
|
44
|
Semba T, Wang X, Xie X, Cohen EN, Reuben JM, Dalby KN, Long JP, Phi LTH, Tripathy D, Ueno NT. Identification of the JNK-Active Triple-Negative Breast Cancer Cluster Associated With an Immunosuppressive Tumor Microenvironment. J Natl Cancer Inst 2022; 114:97-108. [PMID: 34250544 PMCID: PMC8755499 DOI: 10.1093/jnci/djab128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although an immunosuppressive tumor microenvironment (TME) is key for tumor progression, the molecular characteristics associated with the immunosuppressive TME remain unknown in triple-negative breast cancer (TNBC). Our previous functional proteomic study of TNBC tumors identified that C-JUN N-terminal kinase (JNK) pathway-related molecules were enriched in a cluster associated with the inflammatory pathway. However, the role of the JNK pathway in the TNBC TME is still unclear. METHODS Transcriptomic analysis was conducted using The Cancer Genome Atlas datasets. The effect of JNK-IN-8, a covalent pan-JNK inhibitor, on TNBC tumor growth, lung metastasis, and the TME was measured in TNBC syngeneic mouse models (n = 13 per group). Tumor (n = 43) or serum (n = 46) samples from TNBC patients were analyzed using multiplex immunohistochemistry or Luminex assay. All statistical tests were 2-sided. RESULTS CIBERSORT analysis revealed that TNBC patients with high phosphorylated JNK level (n = 47) had more regulatory T cell (Treg) infiltration than those with a low phosphorylated JNK level (n = 47) (P = .02). Inhibition of JNK signaling statistically significantly reduced tumor growth (P < .001) and tumor-infiltrating Tregs (P = .02) while increasing the infiltration of CD8+ T cells in TNBC mouse models through the reduction of C-C motif ligand 2 (CCL2). Tumor-associated macrophages were the predominant cells secreting CCL2, and inhibition of JNK signaling reduced CCL2 secretion of human primary macrophages. Moreover, in patients with TNBC (n = 43), those with high levels of CCL2+ tumor-associated macrophages had more Treg and less CD8+ T cell infiltration (P = .04), and the serum CCL2 level was associated with poor overall survival (hazard ratio = 2.65, 95% confidence interval = 1.29 to 5.44, P = .008) in TNBC patients (n = 46). CONCLUSIONS The JNK/C-JUN/CCL2 axis contributes to TNBC aggressiveness via forming an immunosuppressive TME and can offer novel therapeutic strategies for TNBC.
Collapse
Affiliation(s)
- Takashi Semba
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuemei Xie
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Evan N Cohen
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kevin N Dalby
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - James P Long
- Department of Biostatistics, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lan Thi Hanh Phi
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Debu Tripathy
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
45
|
Chan JM, Quintanal-Villalonga Á, Gao VR, Xie Y, Allaj V, Chaudhary O, Masilionis I, Egger J, Chow A, Walle T, Mattar M, Yarlagadda DVK, Wang JL, Uddin F, Offin M, Ciampricotti M, Qeriqi B, Bahr A, de Stanchina E, Bhanot UK, Lai WV, Bott MJ, Jones DR, Ruiz A, Baine MK, Li Y, Rekhtman N, Poirier JT, Nawy T, Sen T, Mazutis L, Hollmann TJ, Pe'er D, Rudin CM. Signatures of plasticity, metastasis, and immunosuppression in an atlas of human small cell lung cancer. Cancer Cell 2021; 39:1479-1496.e18. [PMID: 34653364 PMCID: PMC8628860 DOI: 10.1016/j.ccell.2021.09.008] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/26/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022]
Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy that includes subtypes defined by differential expression of ASCL1, NEUROD1, and POU2F3 (SCLC-A, -N, and -P, respectively). To define the heterogeneity of tumors and their associated microenvironments across subtypes, we sequenced 155,098 transcriptomes from 21 human biospecimens, including 54,523 SCLC transcriptomes. We observe greater tumor diversity in SCLC than lung adenocarcinoma, driven by canonical, intermediate, and admixed subtypes. We discover a PLCG2-high SCLC phenotype with stem-like, pro-metastatic features that recurs across subtypes and predicts worse overall survival. SCLC exhibits greater immune sequestration and less immune infiltration than lung adenocarcinoma, and SCLC-N shows less immune infiltrate and greater T cell dysfunction than SCLC-A. We identify a profibrotic, immunosuppressive monocyte/macrophage population in SCLC tumors that is particularly associated with the recurrent, PLCG2-high subpopulation.
Collapse
Affiliation(s)
- Joseph M Chan
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Álvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vianne Ran Gao
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Yubin Xie
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Viola Allaj
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ojasvi Chaudhary
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Ignas Masilionis
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Jacklynn Egger
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrew Chow
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thomas Walle
- Department of Medical Oncology; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Clinical Cooperation Unit Virotherapy; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Marissa Mattar
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dig V K Yarlagadda
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - James L Wang
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| | - Fathema Uddin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Offin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Metamia Ciampricotti
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Besnik Qeriqi
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Amber Bahr
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Umesh K Bhanot
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - W Victoria Lai
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew J Bott
- Thoracic Service, Department of Surgery, Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arvin Ruiz
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marina K Baine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanyun Li
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John T Poirier
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10065, USA
| | - Tal Nawy
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Triparna Sen
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Linas Mazutis
- Institute of Biotechnology, Vilnius University, Vilnius, Lithuania
| | - Travis J Hollmann
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dana Pe'er
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
46
|
Paşcalău AV, Cheregi CD, Mureşan MŞ, Şandor MI, Huniadi CA, Nikin Z, Judea Pusta CT, Bodog FD, Ionescu C, Pop OL. CD4+ CD25+ regulatory T-cells role in tumor microenvironment of the squamous cell carcinoma. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 62:249-253. [PMID: 34609428 PMCID: PMC8597358 DOI: 10.47162/rjme.62.1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Introduction: Squamous cell carcinoma (SCC) is the most common skin cancer with a high rate of death. Different lymphocyte populations play an important role in modulating the immune response in the tumor microenvironment. The increase in the proportion of cluster of differentiation (CD)4+ CD25+ regulatory T-cell (Treg) lymphocytes is associated, in different studies, with the increase of the cell multiplication rate. Aim: To analyze the Treg lymphocyte subpopulations and to correlate the results with the presence of the CD8+ cytotoxic T-cell (Tc) lymphocyte population. Materials and Methods: Sixty primary skin SCC specimens were incubated with anti-CD8 (clone SP57) rabbit monoclonal antibody and anti-CD25 (clone 4C9) mouse monoclonal antibody. Results: The ratio of the intratumoral/peritumoral CD4+ CD25+ forkhead box protein p3 (Foxp3) lymphocytes was 0.46, emphasizing that at tumor margins, where tumor aggressiveness is higher, these lymphocytes subpopulations facilitate tumor progression. The comparative analysis of the tumor microenvironment profile revealed that in the case of intratumoral immune response, the number of Tc-type lymphocytes (CD8+) was 3.34 times higher compared to Treg lymphocytes (p<0001). In the peritumoral area, the number of Tc lymphocytes was 5.05 times higher compared to Treg lymphocytes (p<0001). Conclusions: Treg lymphocytes inhibition may cause the suppression of the antitumoral cell immune response in the tumor environment. We believe that Treg lymphocytes should represent a focus of interest for a new personalized therapy. New studies are needed to better understand the immune response in the tumor microenvironment.
Collapse
Affiliation(s)
- Andrei Vasile Paşcalău
- Department of Surgical Disciplines, Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Romania; ,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chamseddine AN, Assi T, Mir O, Chouaib S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol Ther 2021; 231:107986. [PMID: 34481812 DOI: 10.1016/j.pharmthera.2021.107986] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) plasticity and diversity are both essential hallmarks of the monocyte-macrophage lineage and the tumor-derived inflammation. TAM exemplify the perfect adaptable cell with dynamic phenotypic modifications that reflect changes in their functional polarization status. Under several tumor microenvironment (TME)-related cues, TAM shift their polarization, hence promoting or halting cancer progression. Immune checkpoint inhibitors (ICI) displayed unprecedented clinical responses in various refractory cancers; but only approximately a third of patients experienced durable responses. It is, therefore, crucial to enhance the response rate of immunotherapy. Several mechanisms of resistance to ICI have been elucidated including TAM role with its essential immunosuppressive functions that reduce both anti-tumor immunity and the subsequent ICI efficacy. In the past few years, thorough research has led to a better understanding of TAM biology and innovative approaches can now be adapted through targeting macrophages' recruitment axis as well as TAM activation and polarization status within the TME. Some of these therapeutic strategies are currently being evaluated in several clinical trials in association with ICI agents. This combination between TAM modulation and ICI allows targeting TAM intrinsic immunosuppressive functions and tumor-promoting factors as well as overcoming ICI resistance. Hence, such strategies, with a better understanding of the mechanisms driving TAM modulation, may have the potential to optimize ICI efficacy.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Biostatistics and Epidemiology, CESP INSERM U1018, OncoStat, Gustave Roussy, F-94805, Villejuif, France.
| | - Tarek Assi
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Pharmacology, Gustave Roussy, F-94805, Villejuif, France; Department of Ambulatory Care, Gustave Roussy, F-94805, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
48
|
Chen B, Zhang G, Lai J, Xiao W, Li X, Li C, Mok H, Li K, Wang Y, Cao L, Jia M, Ren C, Wen L, Wei G, Lin J, Li Y, Zhang Y, Chen X, Wu X, Zhang H, Li M, Liu J, Balch CM, Liao N. Genetic and immune characteristics of sentinel lymph node metastases and multiple lymph node metastases compared to their matched primary breast tumours. EBioMedicine 2021; 71:103542. [PMID: 34454403 PMCID: PMC8399410 DOI: 10.1016/j.ebiom.2021.103542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Patients with breast cancer presenting with single lymph node metastasis (from a sentinel node) experience prolonged survival compared to patients with multiple lymph node metastases (≥3). However, little information is available on the genetic and immunological characteristics of breast cancer metastases within the regional lymph nodes as they progress from the sentinel lymph node (SLN) downstream to multiple regional lymph nodes (MLNs). METHODS Genomic profiling was performed using a next-generation sequencing panel covering 520 cancer-related genes in the primary tumour and metastatic lymph nodes of 157 female patients with breast cancer. We included primary tumours, metastatic lymph nodes and adjacent clinically normal lymph nodes (20 patients from the SLN group and 28 patients from the MLNs group) in the whole transcriptome analysis. FINDINGS The downstream metastatic lymph nodes (P = 0.029) and the primary breast tumours (P = 0.011) had a higher frequency of PIK3CA mutations compared to the SLN metastasis. We identified a distinct group of 14 mutations from single sentinel node metastasis and a different group of 15 mutations from multiple nodal metastases. Only 4 distinct mutations (PIK3CA, CDK4, NFKBIA and CDKN1B) were conserved in metastases from both lymph node settings. The tumour mutational burden (TMB) was significantly lower in single nodal metastasis compared to the paired primary breast cancer (P = 0.0021), while the decline in TMB did not reach statistical significance in the MLNs group (P = 0.083). In the gene set enrichment analysis, we identified 4 upregulated signatures in both primary tumour and nodal metastases from the MLNs group, including 3 Epithelial-mesenchymal transition(EMT) signatures and 1 angiogenesis signature. Both the CD8/Treg ratio and the CD8/EMT ratio were significantly higher in adjacent normal lymph nodes from patients with a single metastasis in the SLN compared with samples from the MLNs group (P = 0.045 and P = 0.023, respectively). This suggests that the immune defence from the MLNs patients might have a less favourable microenvironment, thus permitting multiple lymph nodes metastasis. INTERPRETATION Single lymph node metastases and multiple lymph node metastases have significant differences in their molecular profiles and immune profiles. The findings are associated with more aggressive tumour characteristics and less favourable immune charactoristics in patients with multiple nodal metastases compared to those with a single metastasis in the sentinel node. FUNDING This work was supported by funds from High-level Hospital Construction Project (DFJH201921), the National Natural Science Foundation of China (81902828 and 82002928), the Fundamental Research Funds for the Central Universities (y2syD2192230), and the Medical Scientific Research Foundation of Guangdong Province (B2019039).
Collapse
Affiliation(s)
- Bo Chen
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Guochun Zhang
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China;; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China;; Shantou University Medical College, Shantou, Guangdong, China
| | - Jianguo Lai
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Weikai Xiao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xuerui Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Cheukfai Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hsiaopei Mok
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Kai Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yulei Wang
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Li Cao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Minghan Jia
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Chongyang Ren
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lingzhu Wen
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Guangnan Wei
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Jiali Lin
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yingzi Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; Shantou University Medical College, Shantou, Guangdong, China
| | - Yuchen Zhang
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Xiaoqing Chen
- Department of Breast Surgical Oncology, Foshan Maternity and Children's Healthcare Hospital Affiliated to Southern Medical University, Foshan, Guangdong, China
| | - Xueying Wu
- Genecast Biotechnology Co., Ltd; Beijing, China
| | - Henghui Zhang
- Genecast Biotechnology Co., Ltd; Beijing, China;; Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Min Li
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Jing Liu
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Charles M Balch
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; School of Medicine, South China University of Technology, 106 Zhongshan Er Road, Guangzhou 510080, Guangdong, China;; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China;; Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
49
|
Abken H. IL-2 "Backpacking" Invigorates Treg Cells to Prevent Allograft Rejection. Transplantation 2021; 105:1394-1395. [PMID: 34157716 DOI: 10.1097/tp.0000000000003614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Hinrich Abken
- RCI, Regensburg Center for Interventional Immunology, Department for Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
50
|
Hu Y, Paris S, Barsoumian H, Abana CO, He K, Wasley M, Younes AI, Masrorpour F, Chen D, Yang L, Dunn JD, Zhang J, Gandhi S, Nguyen QN, Cortez MA, Welsh J. Radiation Therapy Enhanced by NBTXR3 Nanoparticles Overcomes Anti-PD1 Resistance and Evokes Abscopal Effects. Int J Radiat Oncol Biol Phys 2021; 111:647-657. [PMID: 34242713 DOI: 10.1016/j.ijrobp.2021.06.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/10/2021] [Accepted: 06/30/2021] [Indexed: 11/15/2022]
Abstract
PURPOSE Radiation combined with PD1 blockade offers significant treatment benefits in several tumor types; however, anti-PD1 resistance precludes such benefits in many cases. Here we attempted to overcome anti-PD1 resistance by combining localized radiation with a radioenhancing nanoparticle (NBTXR3) and systemic anti-PD1 treatment to achieve abscopal effects in an anti-PD1-resistant mouse model of lung cancer. METHODS AND MATERIALS Female 129Sv/Ev mice were inoculated with 344SQ anti-PD1-resistant (344SQR) or anti-PD1-sensitive (344SQP) metastatic lung cancer cells in the right leg on day 0 ("primary" tumor) and the left leg on day 4 ("secondary" tumor). Primary tumors were injected intratumorally with NBTXR3 on day 7 and were irradiated with 12 Gy on days 8, 9, and 10. Mice were given 6 intraperitoneal injections of anti-PD1. T cell receptor repertoire was analyzed in tumor samples with RNA sequencing, infiltration of CD8 T cells with immunohistochemical staining, and activities of various immune pathways with NanoString analysis. RESULTS The triple combination of NBTXR3 with localized radiation and systemic anti-PD1 significantly delayed the growth of both irradiated and unirradiated tumors in both 344SQP and 344SQR tumor models. NBTXR3 remodeled the immune microenvironment of unirradiated tumors by triggering the activation of various immune pathways, increasing the number of CD8+ T cells, and modifying the T cell receptor repertoire in the 344SQR tumor model. CONCLUSIONS The ability of NBTXR3 to evoke significant abscopal effects in both anti-PD1-sensitive and anti-PD1-resistant lung cancers could open the possibility of its use for treating patients with metastatic lung cancer regardless of sensitivity (or resistance) to immunotherapies.
Collapse
Affiliation(s)
- Yun Hu
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Sébastien Paris
- Department of Translational Science, Nanobiotix, Paris, France
| | | | - Chike O Abana
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Kewen He
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Mark Wasley
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | - Fatemeh Masrorpour
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liangpeng Yang
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Joe Dan Dunn
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Jie Zhang
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Saumil Gandhi
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | - James Welsh
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|