1
|
Zeng S, Xing S, Zhang Y, Wang H, Liu Q. Nano-Bacillus Calmette-Guérin immunotherapies for improved bladder cancer treatment. J Zhejiang Univ Sci B 2024; 25:557-567. [PMID: 39011676 PMCID: PMC11254686 DOI: 10.1631/jzus.b2300392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/29/2023] [Indexed: 07/13/2024]
Abstract
Cancer immunotherapy has rapidly become the fourth mainstream treatment alternative after surgery, radiotherapy, and chemotherapy, with some promising results. It aims to kill tumor cells by mobilizing or stimulating cytotoxic immune cells. However, the clinical applications of tumor immunotherapies are limited owing to a lack of adequate delivery pathways and high toxicity. Recently, nanomaterials and genetic engineering have shown great potential in overcoming these limitations by protecting the delivery of antigens, activating targeted T cells, modulating the immunosuppressive tumor microenvironment, and improving the treatment efficacy. Bacillus Calmette-Guérin (BCG) is a live attenuated Mycobacterium bovis vaccine used to prevent tuberculosis, which was first reported to have antitumor activity in 1927. BCG therapy can activate the immune system by inducing various cytokines and chemokines, and its specific immune and inflammatory responses exert antitumor effects. BCG was first used during the 1970s as an intravesical treatment agent for bladder cancer, which effectively improved immune antitumor activity and prevented tumor recurrence. More recently, nano-BCG and genetically engineered BCG have been proposed as treatment alternatives for bladder cancer due to their ability to induce stronger and more stable immune responses. In this study, we outline the development of nano-BCG and genetically engineered BCG for bladder cancer immunotherapy and review their potential and associated challenges.
Collapse
Affiliation(s)
- Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Shaoqiang Xing
- Department of Urology, First Central Clinical College, Tianjin Medical University, Tianjin 300192, China
| | - Yifei Zhang
- Department of Urology, First Central Clinical College, Tianjin Medical University, Tianjin 300192, China
| | - Haifeng Wang
- Department of Urology, Tianjin First Central Hospital, Tianjin 300192, China.
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin 300192, China.
| |
Collapse
|
2
|
Tamalunas A, Aydogdu C, Unterrainer LM, Schott M, Rodler S, Ledderose S, Schulz GB, Stief CG, Casuscelli J. The Vanishing Clinical Value of PD-L1 Status as a Predictive Biomarker in the First-Line Treatment of Urothelial Carcinoma of the Bladder. Cancers (Basel) 2024; 16:1536. [PMID: 38672618 PMCID: PMC11049370 DOI: 10.3390/cancers16081536] [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: 03/21/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Our study endeavors to elucidate the clinical implications of PD-L1 positivity in individuals afflicted with advanced urothelial carcinoma of the bladder (UCB). METHODS Patients with advanced UCB were prospectively enrolled following a radical cystectomy (RC) performed within January 2017 to December 2022 at our tertiary referral center. The clinical outcome, defined as the progression-free survival (PFS) and overall survival (OS) on systemic treatment, was analyzed using an χ2-test, Mann-Whitney U-test, the Kaplan-Meier method, and a log-rank test. RESULTS A total of 648 patients were included following an RC performed within January 2017 to December 2022. Their PD-L1 status was analyzed with the primary PD-L1-specific antibody (clone SP263, Ventana) and defined both by the CPS and IC-score in 282 patients (43.5%) with a high risk (pT3-pT4 and/or lymph node involvement) or metastatic UCB. While the median PFS was significantly prolonged 5-fold in PD-L1+ patients, we found no difference in OS, regardless of PD-L1 status, or treatment regimen. CONCLUSIONS While PD-L1 positivity indicates prolonged PFS, the presence of PD-L1 does not influence OS rates, suggesting its limited usefulness as a prognostic biomarker in bladder cancer. However, the positive correlation between an PD-L1 status and a sustained response to ICI treatments indicates its potential role as a predictive biomarker. Further research is required to understand how the predictive value of PD-L1 positivity may extend to the use of ICIs in combination with antibody-drug conjugates.
Collapse
Affiliation(s)
- Alexander Tamalunas
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
| | - Can Aydogdu
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Melanie Schott
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
| | - Severin Rodler
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
| | - Stephan Ledderose
- Department of Pathology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Gerald B. Schulz
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
| | - Christian G. Stief
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
| | - Jozefina Casuscelli
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (C.A.); (M.S.); (S.R.); (G.B.S.); (C.G.S.); (J.C.)
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital, 81377 Munich, Germany
| |
Collapse
|
3
|
Stolfo JB, Motta ACDA. Density of high endothelial venules and PDL-1 expression: relationship with tumor-infiltrating lymphocytes in primary cutaneous melanomas. AN ACAD BRAS CIENC 2024; 96:e20230441. [PMID: 38511744 DOI: 10.1590/0001-3765202420230441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/17/2023] [Indexed: 03/22/2024] Open
Abstract
Studies have highlighted melanoma immunogenicity, and the prognostic importance of tumor infiltrating lymphocytes (TILs) and mechanisms of tumor immune evasion, such as hyperexpression of programmed cell death ligand 1 (PDL-1). High endothelial venules (HEV) are specialized blood vessels that can facilitate the lymphocytes migration to the tumor. Here we evaluate the association of HEV density and PDL-1 expression in primary cutaneous melanomas with the presence and degree of TILs and with other clinicopathological variables (age, sex, tumor location, melanoma histological type, Breslow thickness, ulceration, regression signs, mitotic index). HEV density and PDL-1 expression were assessed immunohistochemically in 78 melanoma cases, using a specific antibody, and were detected in 59% and 76% of these, respectively. Positive associations were identified between HEV density and PDL-1 expression with the presence and degree of lymphocytic infiltration, melanoma histological type and ulceration presence. No correlation was found between HEV density and PDL-1 expression. Our findings confirm the HEV role in the recruitment and facilitation of lymphocyte transport in cutaneous melanomas, where HEV density is strongly associated with the degree of TILs. Additionally, PDL-1 hyperexpression suggests a possible mechanism of tumor immune evasion, which may lead to inactivation and reduction of the tumor lymphocytes number.
Collapse
Affiliation(s)
- Josiane B Stolfo
- Programa de Pós-Graduação em Bioexperimentação, Universidade de Passo Fundo, Escola de Ciências Agrárias, Inovação e Negócios, Campus I, BR 285, Km 171, São José, 99001-970 Passo Fundo, RS, Brazil
| | - Adriana C DA Motta
- Programa de Pós-Graduação em Bioexperimentação, Universidade de Passo Fundo, Escola de Ciências Agrárias, Inovação e Negócios, Campus I, BR 285, Km 171, São José, 99001-970 Passo Fundo, RS, Brazil
| |
Collapse
|
4
|
Maher NG, Vergara IA, Long GV, Scolyer RA. Prognostic and predictive biomarkers in melanoma. Pathology 2024; 56:259-273. [PMID: 38245478 DOI: 10.1016/j.pathol.2023.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/20/2023] [Indexed: 01/22/2024]
Abstract
Biomarkers help to inform the clinical management of patients with melanoma. For patients with clinically localised primary melanoma, biomarkers can help to predict post-surgical outcome (including via the use of risk prediction tools), better select patients for sentinel lymph node biopsy, and tailor catch-all follow-up protocols to the individual. Systemic drug treatments, including immune checkpoint inhibitor (ICI) therapies and BRAF-targeted therapies, have radically improved the prognosis of metastatic (stage III and IV) cutaneous melanoma patients, and also shown benefit in the earlier setting of stage IIB/C primary melanoma. Unfortunately, a response is far from guaranteed. Here, we review clinically relevant, established, and emerging, prognostic, and predictive pathological biomarkers that refine clinical decision-making in primary and metastatic melanoma patients. Gene expression profile assays and nomograms are emerging tools for prognostication and sentinel lymph node risk prediction in primary melanoma patients. Biomarkers incorporated into clinical practice guidelines include BRAF V600 mutations for the use of targeted therapies in metastatic cutaneous melanoma, and the HLA-A∗02:01 allele for the use of a bispecific fusion protein in metastatic uveal melanoma. Several predictive biomarkers have been proposed for ICI therapies but have not been incorporated into Australian clinical practice guidelines. Further research, validation, and assessment of clinical utility is required before more prognostic and predictive biomarkers are fluidly integrated into routine care.
Collapse
Affiliation(s)
- Nigel G Maher
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ismael A Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Fiorentino V, Pizzimenti C, Franchina M, Pepe L, Russotto F, Tralongo P, Micali MG, Militi GB, Lentini M. Programmed Cell Death Ligand 1 Immunohistochemical Expression and Cutaneous Melanoma: A Controversial Relationship. Int J Mol Sci 2024; 25:676. [PMID: 38203846 PMCID: PMC10779806 DOI: 10.3390/ijms25010676] [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: 10/09/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Cutaneous melanoma (CM) is traditionally considered one of the most "immunogenic" tumors, eliciting a high immune response. However, despite the presence of tumor-infiltrating lymphocytes (TILs), melanoma cells use strategies to suppress antitumor immunity and avoid being eliminated by immune surveillance. The PD-1 (programmed death-1)/PD-L1 (programmed death-ligand 1) axis is a well-known immune escape system adopted by neoplastic cells. Therefore, immunotherapy with PD-1 and PD-L1 inhibitors is quickly becoming the main treatment approach for metastatic melanoma patients. However, the clinical utility of PD-L1 expression assessment in CM is controversial, and the interpretation of PD-L1 scores in clinical practice is still a matter of debate. Nonetheless, the recent literature data show that by adopting specific PD-L1 assessment methods in melanoma samples, a correlation between the expression of such a biomarker and a positive response to PD-1-based immunotherapy can be seen. Our review aims to describe the state-of-the-art knowledge regarding the prognostic and predictive role of PD-L1 expression in CM while also referring to possible biological explanations for the variability in its expressions and related treatment responses.
Collapse
Affiliation(s)
- Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Ludovica Pepe
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Fernanda Russotto
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Pietro Tralongo
- Department of Women, Children and Public Health Sciences, Catholic University of the Sacred Heart, Agostino Gemelli IRCCS University Hospital Foundation, 00168 Rome, Italy;
| | - Marina Gloria Micali
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Gaetano Basilio Militi
- Department of Sciences for Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, 90133 Palermo, Italy;
| | - Maria Lentini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| |
Collapse
|
6
|
Cobankent Aytekin E, Unal B, Bassorgun CI, Ozkan O. Clinicopathologic Evaluation of CD80, CD86, and PD-L1 Expressions with Immunohistochemical Methods in Malignant Melanoma Patients. Turk Patoloji Derg 2024; 40:16-26. [PMID: 37614091 PMCID: PMC10823788 DOI: 10.5146/tjpath.2023.01608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/27/2023] [Indexed: 08/25/2023] Open
Abstract
OBJECTIVE Diagnostic and prognostic biomarkers for malignant melanoma are crucial for treatment and for developing targeted therapies. Malignant melanoma is a highly immunogenic tumor, and its regression, treatment, and prognostic evaluation are directly related to escape from immune destruction. Therefore, we aimed to determine the expression levels of CD80, CD86, and PD -L1 in malignant melanoma tissue samples by immunohistochemistry and to investigate the possible relationship between these proteins and the clinicopathological features in this study. MATERIAL AND METHODS Hematoxylin and eosin staining and immunohistochemical staining for CD80, CD86, and PD-L1 were evaluated for clinical data, survival, prognosis, tumor location, malignant melanoma subtypes, tumor size, and prognostic findings. RESULTS Higher survival rates were observed in patients with lower PD-L1 staining scores in the tumor. The 5-year survival was higher in patients with CD80-positive and CD86-positive biopsies. Mortality was lower in superficial spreading melanoma and Lentigo maligna melanoma types, whereas staining positivity of CD80 and CD86 was higher. Furthermore, a relationship between clinical stage and Breslow thickness ( < 2mm/≥2mm), tumor ulceration, lymph node metastasis, and CD80 and CD86 expression was also identified. CONCLUSION Our findings suggest that PD-L1, CD80, and CD86 expression are essential in malignant melanoma and could be used as prognostic markers.
Collapse
Affiliation(s)
| | - Betul Unal
- Department of Pathology, Akdeniz University, Faculty of Medicine, Antalya, Turkey
| | | | - Ozlenen Ozkan
- Department of Plastic and Reconstructive Surgery, Akdeniz University, Faculty of Medicine, Antalya, Turkey
| |
Collapse
|
7
|
Cheng L, Yu J, Hao T, Wang W, Wei M, Li G. Advances in Polymeric Micelles: Responsive and Targeting Approaches for Cancer Immunotherapy in the Tumor Microenvironment. Pharmaceutics 2023; 15:2622. [PMID: 38004600 PMCID: PMC10675796 DOI: 10.3390/pharmaceutics15112622] [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: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
In recent years, to treat a diverse array of cancer forms, considerable advancements have been achieved in the field of cancer immunotherapies. However, these therapies encounter multiple challenges in clinical practice, such as high immune-mediated toxicity, insufficient accumulation in cancer tissues, and undesired off-target reactions. To tackle these limitations and enhance bioavailability, polymer micelles present potential solutions by enabling precise drug delivery to the target site, thus amplifying the effectiveness of immunotherapy. This review article offers an extensive survey of recent progress in cancer immunotherapy strategies utilizing micelles. These strategies include responsive and remodeling approaches to the tumor microenvironment (TME), modulation of immunosuppressive cells within the TME, enhancement of immune checkpoint inhibitors, utilization of cancer vaccine platforms, modulation of antigen presentation, manipulation of engineered T cells, and targeting other components of the TME. Subsequently, we delve into the present state and constraints linked to the clinical utilization of polymeric micelles. Collectively, polymer micelles demonstrate excellent prospects in tumor immunotherapy by effectively addressing the challenges associated with conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Lichun Cheng
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Jiankun Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Tangna Hao
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Wenshuo Wang
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Guiru Li
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| |
Collapse
|
8
|
Warli SM, Prapiska FF, Siregar DIS, Seja IA. Tumor Markers as Predictors of Acute Kidney Injury Incidence and Staging of the Muscle-Invasive Bladder Cancer Receiving Chemoradiation Therapy. World J Oncol 2023; 14:423-429. [PMID: 37869246 PMCID: PMC10588499 DOI: 10.14740/wjon1676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 10/24/2023] Open
Abstract
Background Bladder cancer, as one of types of cancers within the urinary tract, is associated with a greater risk of acute kidney injury (AKI), resulting in a poorer prognosis, discontinuation of effective oncological treatments, longer hospitalization, and higher expenses. There is no discussion yet on tumor markers in bladder cancer. With the revolutionary advances in bladder cancer molecular subtyping over the past decade, the presence of tumor markers to assess the staging of bladder cancer has yet to be discussed. In this study, we intended to assess the relationship between tumor markers and incidence of AKI, also between tumor markers and the cancer staging. Methods This retrospective cross-sectional study utilized secondary data from 26 medical records of patients diagnosed with bladder cancer at the Adam Malik and Universitas Sumatera Utara Hospital between 2021and 2022. This study included all patients with bladder cancer who met the inclusion criteria. Continuous variables were reported as mean (standard deviation (SD)) and examined using an independent t-test. Categorical variables were reported as proportions, examined using Chi-square or Fisher's exact test. Pre- and post-tumor marker data were evaluated with dependent sample t-test for normal variance data, and Wilcoxon test for data with atypical distribution. P values were set at 0.05. Results CD44 (P = 0.003) and programmed cell death 1 (PD-1) (P = 0.030) were the only significant markers in their pre- and post-chemoradiation states among the four investigated tumor markers in this study. Meanwhile, PD-1 tumor marker levels were only found to be significant between AKI and pre-chemoradiation (P = 0.011). Even though the multivariate study of tumor staging did not show any statistical significance, both tumor markers CD44 and PD-1 showed a significant effect on the incidence of acute renal damage (P = 0.034). Conclusions Pre-chemoradiation PD-1 tumor markers showed promise as good predictive indicators for staging and AKI incidence in muscle-invasive bladder cancer patients undergoing chemoradiation therapy.
Collapse
Affiliation(s)
- Syah Mirsya Warli
- Division of Urology, Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara - Haji Adam Malik General Hospital, Medan, Indonesia
- Department of Urology, Faculty of Medicine, Universitas Sumatera Utara Hospital - Universitas Sumatera Utara, Medan, Indonesia
| | - Fauriski Febrian Prapiska
- Department of Urology, Faculty of Medicine, Universitas Sumatera Utara Hospital - Universitas Sumatera Utara, Medan, Indonesia
| | - Dewi Indah Sari Siregar
- Department of Clinical Pathology, Faculty of Medicine, Universitas Sumatera Utara Hospital - Universitas Sumatera Utara, Medan, Indonesia
| | - Ilham Ari Seja
- Department of Urology, Faculty of Medicine, Universitas Indonesia - Haji Adam Malik General Hospital, Medan, Indonesia
| |
Collapse
|
9
|
Cazzato G, Lettini T, Colagrande A, Trilli I, Ambrogio F, Laface C, Parente P, Maiorano E, Ingravallo G. Immunohistochemical Expression of Programmed Cell Death Ligand 1 (PD-L1) in Human Cutaneous Malignant Melanoma: A Narrative Review with Historical Perspectives. Genes (Basel) 2023; 14:1252. [PMID: 37372432 DOI: 10.3390/genes14061252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is the primary ligand of the receptor programmed death-1 (PD-1) which is constitutively expressed or activated in myeloid, lymphoid (T, B and NK), normal epithelial cells, and cancer. The PD-1/PD-L1 interaction is crucial for the physiological development of immunological tolerance but also in the development of the cancer. Among these, malignant melanoma represents a tumour in which the immunohistochemical expression of PD-L1 is important to guide future therapeutic choices based on the presence/absence of expression. Various clones have been used over time for immunohistochemical determination, and different results and heterogeneity remain among the various studies in the literature. We perform a narrative review of the present studies in order to discuss and take stock of what certain achievements have been made in this field, what challenges remain, and what possible solutions can be found.
Collapse
Affiliation(s)
- Gerardo Cazzato
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Teresa Lettini
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Anna Colagrande
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Irma Trilli
- Odontomatostologic Clinic, Department of Innovative Technologies in Medicine and Dentistry, University of Chieti, 66100 Chieti, Italy
| | - Francesca Ambrogio
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, BR, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy
| | - Eugenio Maiorano
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| | - Giuseppe Ingravallo
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy
| |
Collapse
|
10
|
Bahreyni A, Mohamud Y, Luo H. Recent advancements in immunotherapy of melanoma using nanotechnology-based strategies. Biomed Pharmacother 2023; 159:114243. [PMID: 36641926 DOI: 10.1016/j.biopha.2023.114243] [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: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Melanoma is a malignant tumor that accounts for the deadliest form of skin cancers. Despite the significant efforts made recently for development of immunotherapeutic strategies including using immune checkpoint inhibitors and cancer vaccines, the clinical outcomes are unsatisfying. Different factors affect efficient cancer immunotherapy such as side-effects, immunosuppressive tumor microenvironment, and tumor heterogeneity. In the past decades, various nanotechnology-based approaches have been developed to enhance the efficacy of cancer immunotherapy, in addition to diminishing the toxicity associated with it. Several studies have shown that proper application of nanomaterials can revolutionize the outcome of immunotherapy in diverse melanoma models. This review summarizes the recent advancement in the integration of nanotechnology and cancer immunotherapy in melanoma treatment. The importance of nanomaterials and their therapeutic advantages for patients with melanoma are also discussed.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
11
|
Xin X, Zhou Y, Li J, Zhang K, Qin C, Yin L. CXCL10-coronated thermosensitive "stealth" liposomes for sequential chemoimmunotherapy in melanoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102634. [PMID: 36462759 DOI: 10.1016/j.nano.2022.102634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/05/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
The interplay of liposome-protein corona hinders the clinical application of liposomes due to active macrophage sequestration and rapid plasma clearance. Here we showed that, CXCL10 as a therapeutic protein was coronated the thermosensitive liposomes to form stealth-like nanocarriers (CXCL10/TSLs). Decoration of the corona layer of CXCL10/TSLs by hyaluronic acid conjugated oridonin (ORD/CXCL10/TSLs), overcame the "fluid barrier" built by biological proteins, drastically reduced capture by leukocytes in whole blood, allowed the specific targeting of tumor sites. Multifunctional medicine ORD/CXCL10/TSLs with hyperthermia drove the sustained cytokine-CXCL10 inflammatory loop to switch macrophage phenotype to M1-like, expand tumor-infiltrating natural killer cells and induce intratumoral levels of interferon-γ. Oridonin synergized with CXCL10 during ORD/CXCL10/TSLs treatment, downregulated PI3K/AKT and Raf/MEK signaling for M1-like polarization and migration inhibition. Furthermore, ORD/CXCL10/TSLs potently synergized with anti-PD-L1 antibody in mice bearing metastatic melanoma, induced sustained immunological memory and controlled metastatic spread.
Collapse
Affiliation(s)
- Xiaofei Xin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Yong Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jingjing Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Kai Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chao Qin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, China; State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
PD-L1, CD4+, and CD8+ Tumor-Infiltrating Lymphocytes (TILs) Expression Profiles in Melanoma Tumor Microenvironment Cells. J Pers Med 2023; 13:jpm13020221. [PMID: 36836455 PMCID: PMC9965691 DOI: 10.3390/jpm13020221] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
(1) Background: Because melanoma is an aggressive tumor with an unfavorable prognosis, we aimed to characterize the PD-L1 expression in melanomas in association with T cell infiltrates because PD-1/PD-L1 blockade represents the target in treating melanoma strategy. (2) Methods: The immunohistochemical manual quantitative methods of PD-L1, CD4, and CD8 TILs were performed in melanoma tumor microenvironment cells. (3) Results: Most of the PD-L1 positive, expressing tumors, have a moderate score of CD4+ TILs and CD8+TILs (5-50% of tumor area) in tumoral melanoma environment cells. The PD-L1 expression in TILs was correlated with different degrees of lymphocytic infiltration described by the Clark system (X2 = 8.383, p = 0.020). PD-L1 expression was observed often in melanoma cases, with more than 2-4 mm of Breslow tumor thickness being the associated parameters (X2 = 9.933, p = 0.014). (4) Conclusions: PD-L1 expression represents a predictive biomarker with very good accuracy for discriminating the presence or absence of malign tumoral melanoma cells. PD-L1 expression was an independent predictor of good prognosis in patients with melanomas.
Collapse
|
13
|
Li D, Ma L, Bao J, Cao L, Min W. PD-L1 Biomolecules Associated with Clinical Features in Non-Melanoma Skin Cancer. Clin Cosmet Investig Dermatol 2023; 16:1-8. [PMID: 36628329 PMCID: PMC9826606 DOI: 10.2147/ccid.s383481] [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: 08/04/2022] [Accepted: 11/18/2022] [Indexed: 01/05/2023]
Abstract
Background Increasing evidence has indicated that several B7 family members play critical roles in the progress of many cancers. However, the clinical significance of the B7 family in cutaneous squamous cell carcinoma (cSCC) is still elusive. The purpose of this study is to investigate the potential role of B7-H1 biomolecules (PD-L1) in regulating the tumorigenesis and progression of cSCC, the most common non-melanoma skin cancer. Methods We collected transcriptome data of cSCC patients from TCGA databases (n = 496) and subjected the transcription data to bioinformatical analysis. Differential expression of B7-H1 genes with a grade-dependent pattern was identified. We collected paraffin sections of skin squamous carcinoma and analyzed by immunohistochemical staining. We further examined the PD-L1 levels of CD14+ cells in peripheral blood of each cSCC patient and normal subjects by flow cytometry. Results It was found that higher expression of PD-L1 was associated with poor prognosis of cSCC patients and shorter overall survival. These observations were further verified in the clinical paraffin sections and in peripheral blood T cells. Conclusion Our study reveals that PD-L1 is a potential prognostic marker in clinical prognosis for cSCC patients and could be valuable for cSCC treatment.
Collapse
Affiliation(s)
- Dan Li
- Department of Dermatology, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Liwen Ma
- Department of Dermatology, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Jun Bao
- Department of Dermatology, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Lei Cao
- Clinical Immunology Laboratory, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
| | - Wei Min
- Department of Dermatology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China,Correspondence: Wei Min, Email
| |
Collapse
|
14
|
Wang JZ, Nassiri F, Bi L, Zadeh G. Immune Profiling of Meningiomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1416:189-198. [PMID: 37432628 DOI: 10.1007/978-3-031-29750-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Though meningiomas are generally regarded as benign tumors, there is increasing awareness of a large group of meningiomas that are biologically aggressive and refractory to the current standards of care treatment modalities. Coinciding with this has been increasing recognition of the important that the immune system plays in mediating tumor growth and response to therapy. To address this point, immunotherapy has been leveraged for several other cancers such as lung, melanoma, and recently glioblastoma in the context of clinical trials. However, first deciphering the immune composition of meningiomas is essential in order to determine the feasibility of similar therapies for these tumors. Here in this chapter, we review recent updates on characterizing the immune microenvironment of meningiomas and identify potential immunological targets that hold promise for future immunotherapy trials.
Collapse
Affiliation(s)
- Justin Z Wang
- Division of Neurosurgery, Department of Surgery, The University of Toronto, Toronto, ON, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, Department of Surgery, The University of Toronto, Toronto, ON, Canada.
| | - Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, The University of Toronto, Toronto, ON, Canada
- Department of Neurosurgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
The Association of CD8+ Cytotoxic T Cells and Granzyme B+ Lymphocytes with Immunosuppressive Factors, Tumor Stage and Prognosis in Cutaneous Melanoma. Biomedicines 2022; 10:biomedicines10123209. [PMID: 36551965 PMCID: PMC9775436 DOI: 10.3390/biomedicines10123209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
The immunosuppressive tumor microenvironment (TME) consists of suppressive cells producing a variety of immunomodulatory proteins, such as programmed death ligand 1 (PD-L1) and indoleamine-2,3-dioxygenase (IDO). Although granzyme B (GrB) is known to convey the cytolytic activities of CD8+ cytotoxic lymphocytes, it is also expressed by other cells, such as regulatory T and B cells, for immunosuppressive purposes. The role of GrB+ lymphocytes in melanoma has not been examined extensively. In this study, benign, premalignant, and malignant melanocytic tumors were stained immunohistochemically for CD8 and GrB. PD-L1 was also stained from malignant samples that had accompanying clinicopathological data. The association of CD8+ and GrB+ lymphocytes with PD-L1 expression, tumor stage, prognosis, and previously analyzed immunosuppressive factors were evaluated. Our aim was to obtain a more comprehensive perception of the immunosuppressive TME in melanoma. The results show that both CD8+ and GrB+ lymphocytes were more abundant in pT4 compared to pT1 melanomas, and in lymph node metastases compared to primary melanomas. Surprisingly, a low GrB/CD8 ratio was associated with better recurrence-free survival in primary melanomas, which indicates that GrB+ lymphocytes might represent activated immunosuppressive lymphocytes rather than cytotoxic T cells. In the present study, CD8+ lymphocytes associated positively with both tumor and stromal immune cell PD-L1 and IDO expression. In addition, PD-L1+ tumor and stromal immune cells associated positively with IDO+ stromal immune and melanoma cells. The data suggest that IDO and PD-L1 seem to be key immunosuppressive factors in CD8+ lymphocyte-predominant tumors in CM.
Collapse
|
16
|
Najafi S, Majidpoor J, Mortezaee K. The impact of microbiota on PD-1/PD-L1 inhibitor therapy outcomes: A focus on solid tumors. Life Sci 2022; 310:121138. [DOI: 10.1016/j.lfs.2022.121138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/02/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
17
|
Wu CG, Casanova R, Mairinger F, Soltermann A. Lung adenocarcinoma patients with malignant pleural effusions in hot adaptive immunity status have a longer overall survival. Front Oncol 2022; 12:1031094. [PMID: 36267973 PMCID: PMC9577289 DOI: 10.3389/fonc.2022.1031094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Malignant pleural effusion (MPE) is a common complication of lung adenocarcinoma (LADC) which is associated with a dismal prognosis. We investigated the prognostic role of PD-L1 and other immunomodulators expression in the immune compartment of MPE immune composition. MPE cytologic cell blocks of 83 LADC patients were analysed for the mRNA expression of 770 cancer-immune genes by the NanoString nCounter platform. The expression of relevant immune cell lineage markers was validated by immunohistochemistry (IHC) using quantitative pathology. The mRNA immune profiling identified four MPE patient clusters (C). C1/2 (adaptive+, hot) showed better overall survival (OS) than C3/4 (adaptive-, cold). Additionally, cold immunity profiles (adaptive-), C4 (innate+) were associated with worse OS than C3 (innate-). High PD-L1 expression was linked to the regulation of T cell activation and interferon signalling pathways. Genes of pattern recognition receptor and type I interferon signalling pathways were specifically upregulated in the long-survival (≥90 days) patient group. Moreover, immunomodulators were co-activated and highly expressed in hot adaptive immunity patient clusters, whereas CD274 (PD-L1), TNFRSF9 (4-1BB), VEGFA (VEGF-A) and CD276 (B7-H3) were upregulated in the groups referred as cold. The patient cluster, age and PD-L1 expression were independent prognosticators for LADC MPE patients (p-value < 0.05). Our study sheds light on the variances of immune contexture regarding different PD-L1 expression and survival conditions. It revealed four distinct prognostic patient clusters with specific immune cell components and immunomodulator expression profiles, which, collectively, is supportive for future therapeutic and prognosis for cancer management.
Collapse
Affiliation(s)
- Cheng-Guang Wu
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
- *Correspondence: Cheng-Guang Wu, ; Alex Soltermann,
| | - Ruben Casanova
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Alex Soltermann
- Facharzt Foederatio Medicorum Helveticorum (FMH) Pathologie, Pathologie Länggasse, Ittigen, Switzerland
- *Correspondence: Cheng-Guang Wu, ; Alex Soltermann,
| |
Collapse
|
18
|
Rohaan M, Gomez-Eerland R, van den Berg J, Geukes Foppen M, van Zon M, Raud B, Jedema I, Scheij S, de Boer R, Bakker N, van den Broek D, Pronk L, Grijpink-Ongering L, Sari A, Kessels R, van den Haak M, Mallo H, Karger M, van de Wiel B, Zuur C, Duinkerken C, Lalezari F, van Thienen J, Wilgenhof S, Blank C, Beijnen J, Nuijen B, Schumacher T, Haanen J. MART-1 TCR gene-modified peripheral blood T cells for the treatment of metastatic melanoma: a phase I/IIa clinical trial. IMMUNO-ONCOLOGY AND TECHNOLOGY 2022; 15:100089. [PMID: 35865122 PMCID: PMC9293760 DOI: 10.1016/j.iotech.2022.100089] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- M.W. Rohaan
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R. Gomez-Eerland
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J.H. van den Berg
- Biotherapeutics Unit, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M.H. Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M. van Zon
- Biotherapeutics Unit, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - B. Raud
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I. Jedema
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - S. Scheij
- Biotherapeutics Unit, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R. de Boer
- Biotherapeutics Unit, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - N.A.M. Bakker
- Biotherapeutics Unit, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - D. van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L.M. Pronk
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - A. Sari
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R. Kessels
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M. van den Haak
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - H.A. Mallo
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M. Karger
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - B.A. van de Wiel
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C.L. Zuur
- Department of Head and Neck Surgery, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C.W. Duinkerken
- Department of Head and Neck Surgery, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - F. Lalezari
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J.V. van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - S. Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C.U. Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J.H. Beijnen
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - B. Nuijen
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T.N. Schumacher
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - J.B.A.G. Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Correspondence to: Prof. John B. A. G. Haanen, Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands. Tel: 0031-205126979; Fax: 0031-205122572
| |
Collapse
|
19
|
Kuczkiewicz-Siemion O, Sokół K, Puton B, Borkowska A, Szumera-Ciećkiewicz A. The Role of Pathology-Based Methods in Qualitative and Quantitative Approaches to Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14153833. [PMID: 35954496 PMCID: PMC9367614 DOI: 10.3390/cancers14153833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Immunotherapy has become the filar of modern oncological treatment, and programmed death-ligand 1 expression is one of the primary immune markers assessed by pathologists. However, there are still some issues concerning the evaluation of the marker and limited information about the interaction between the tumour and associated immune cells. Recent studies have focused on cancer immunology to try to understand the complex tumour microenvironment, and multiplex imaging methods are more widely used for this purpose. The presented article aims to provide an overall review of a different multiplex in situ method using spectral imaging, supported by automated image-acquisition and software-assisted marker visualisation and interpretation. Multiplex imaging methods could improve the current understanding of complex tumour-microenvironment immunology and could probably help to better match patients to appropriate treatment regimens. Abstract Immune checkpoint inhibitors, including those concerning programmed cell death 1 (PD-1) and its ligand (PD-L1), have revolutionised the cancer therapy approach in the past decade. However, not all patients benefit from immunotherapy equally. The prediction of patient response to this type of therapy is mainly based on conventional immunohistochemistry, which is limited by intraobserver variability, semiquantitative assessment, or single-marker-per-slide evaluation. Multiplex imaging techniques and digital image analysis are powerful tools that could overcome some issues concerning tumour-microenvironment studies. This novel approach to biomarker assessment offers a better understanding of the complicated interactions between tumour cells and their environment. Multiplex labelling enables the detection of multiple markers simultaneously and the exploration of their spatial organisation. Evaluating a variety of immune cell phenotypes and differentiating their subpopulations is possible while preserving tissue histology in most cases. Multiplexing supported by digital pathology could allow pathologists to visualise and understand every cell in a single tissue slide and provide meaning in a complex tumour-microenvironment contexture. This review aims to provide an overview of the different multiplex imaging methods and their application in PD-L1 biomarker assessment. Moreover, we discuss digital imaging techniques, with a focus on slide scanners and software.
Collapse
Affiliation(s)
- Olga Kuczkiewicz-Siemion
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Diagnostic Hematology Department, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland
- Correspondence: (O.K.-S.); (A.S.-C.)
| | - Kamil Sokół
- Diagnostic Hematology Department, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland
| | - Beata Puton
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Aneta Borkowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Anna Szumera-Ciećkiewicz
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Correspondence: (O.K.-S.); (A.S.-C.)
| |
Collapse
|
20
|
Hamed MM, Gouida MS, Abd EL-Aziz SR, EL-Sokkary AM. Evaluation PD-L1, CD8 and CD20 as early predictor and tracking markers for breast cancer (BC) in Egypt. Heliyon 2022; 8:e09474. [PMID: 35647336 PMCID: PMC9136277 DOI: 10.1016/j.heliyon.2022.e09474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/20/2022] [Accepted: 05/13/2022] [Indexed: 11/15/2022] Open
Abstract
Background Breast cancer (BC) is considered as a common type of cancer threatening women throughout the world. Therefore, development of early predication biomarkers for BC got more concern especially for Egyptian females. This study was aimed to evaluate PD-L1, CD8, and CD20 as early prediction breast cancer biomarkers. Methods Flow cytometry (FC), immunohistochemistry (IHC), Western Blot, and q-PCR were used to compare PD-L1, CD20, and CD8 levels in tissues and blood samples of Breast Cancer and controls. Results Blood samples showed a significant increase in PD-L1, CD20, and CD8 compared to controls (p˂0.005). A Significant correlation was shown between PD-L1, CD8, and CD20 in tissue and breast cancer subtypes. Whereas, invasive lobular carcinoma (ILC) was characterized by superior PD-L1 and CD20 levels compared to invasive ductal carcinoma (IDC). FC studies on Blood showed 83% and 45.7% PD-L1 expressions for IDC and ILC, respectively. CD20 in ILC and IDC were 78.2% and 62.5%, respectively. Nevertheless, CD8 was 74.2% for IDC and 67.7% for ILC. Whereas, FC studies for PD-L1, CD20, and CD8 in ILC in tissues gave 34.4%, 30.2% and 35.1%, respectively. In addition, IDC tissue samples showed 16%, 12.5, and 13.5% for PD-L1, CD20, and CD8. The moderate stage of adenocarcinoma caused expression of PD-L1 within inflammatory cells, while expression was within neoplastic glandular cells in late stage. Conclusion PD-L1, CD8, and CD20 are considered as early predictor and tracking markers for breast cancer.
Collapse
Affiliation(s)
- Manar M. Hamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Egypt
| | - Mona S. Gouida
- Genetic Unit, Children Hospital, Mansoura University, Egypt
| | | | - Ahmed M.A. EL-Sokkary
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Egypt
| |
Collapse
|
21
|
Chen M, Wang Y, Wang L, Shen C, Chen C, Lee H. PD-L1 expressed from tumor cells promotes tumor growth and invasion in lung cancer via modulating TGF-β1/SMAD4 expression. Thorac Cancer 2022; 13:1322-1332. [PMID: 35373505 PMCID: PMC9058315 DOI: 10.1111/1759-7714.14388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Programmed death ligand-1 (PD-L1) has a known association with the prognosis of human cancers because of its ability to alter tumor immune surveillance via its interaction with PD-1. We questioned whether expression of PD-L1 in tumor cells could directly promote tumor growth and invasiveness in non-small cell lung cancer (NSCLC). METHODS Real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed to evaluate PD-L1 messenger RNA (mRNA) expression in lung tumors. The prognostic value of PD-L1 mRNA was assessed by Cox regression model. Transcriptional regulation of PD-L1 by human papillomavirus (HPV) 16/18 E6 oncoprotein or by epidermal growth factor receptor (EGFR) mutation in lung cancer cells was examined by Western blot and luciferase reporter assay. The cell growth and invasion were evaluated by colony formation, soft agar growth, and Boyden chamber assay. RESULTS The PD-L1 mRNA levels showed a positive association with HPV 16/18 E6 oncoprotein and with EGFR mutation in 223 surgically resected NSCLC patients. The prognostic significance of PD-L1 was more commonly observed in patients with high PD-L1/E6 positive and high PD-L1/EGFR mutant tumors. Mechanistically, upregulation of PD-L1 transcription by E6 or mutant EGFR occurred largely through the ERK-C/EBPβ-TLR4-NF-κB cascade. PD-L1 promotes the efficacy of colony formation, soft agar growth, and cell invasion. PD-L1 upregulates BAG-1 to reduce transforming growth factor (TGF)-β1 expression, and the decrease in SMAD4 because of TGF-β1 occurs through the p53/microRNA (miR)-224 axis. The decreases in TGF-β1 and SMAD4 are responsible for PD-L1-mediated cell invasiveness. CONCLUSION Induction of PD-L1 by E6 oncoprotein or mutant EGFR through the ERK-C/EBPβ-TLR4-NF-κB cascade may promote tumor growth and invasiveness in NSCLC because of decreasing TGF-β1 and SMAD4 expression.
Collapse
Affiliation(s)
- Ming‐Jenn Chen
- Department of SurgeryChi Mei Medical CenterTainanTaiwan
- Department of Sports Management, College of Leisure and Recreation ManagementChia Nan University of Pharmacy and ScienceTainanTaiwan
| | - Yao‐Chen Wang
- Department of Internal Medicine, Chung Shan Medical University Hospital, School of MedicineChung Shan Medical UniversityTaichungTaiwan
| | - Lee Wang
- Department of Public HealthChung Shan Medical UniversityTaichungTaiwan
| | - Ching‐Ju Shen
- Department of Gynecology and Obstetrics, Kaohsiung Medical University Hospital, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Yi Chen
- Department of SurgeryChung Shan Medical University HospitalTaichungTaiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug DiscoveryTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
22
|
Ernst M, Giubellino A. The Current State of Treatment and Future Directions in Cutaneous Malignant Melanoma. Biomedicines 2022; 10:822. [PMID: 35453572 PMCID: PMC9029866 DOI: 10.3390/biomedicines10040822] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Malignant melanoma is the leading cause of death among cutaneous malignancies. While its incidence is increasing, the most recent cancer statistics show a small but clear decrease in mortality rate. This trend reflects the introduction of novel and more effective therapeutic regimens, including the two cornerstones of melanoma therapy: immunotherapies and targeted therapies. Immunotherapies exploit the highly immunogenic nature of melanoma by modulating and priming the patient's own immune system to attack the tumor. Treatments combining immunotherapies with targeted therapies, which disable the carcinogenic products of mutated cancer cells, have further increased treatment efficacy and durability. Toxicity and resistance, however, remain critical challenges to the field. The present review summarizes past treatments and novel therapeutic interventions and discusses current clinical trials and future directions.
Collapse
Affiliation(s)
| | - Alessio Giubellino
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
23
|
Ushio R, Murakami S, Saito H. Predictive Markers for Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. J Clin Med 2022; 11:1855. [PMID: 35407463 PMCID: PMC9000007 DOI: 10.3390/jcm11071855] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically improved the outcomes of non-small cell lung cancer patients and have increased the possibility of long-term survival. However, few patients benefit from ICIs, and no predictive biomarkers other than tumor programmed cell death ligand 1 (PD-L1) expression have been established. Hence, the identification of biomarkers is an urgent issue. This review outlines the current understanding of predictive markers for the efficacy of ICIs, including PD-L1, tumor mutation burden, DNA mismatch repair deficiency, microsatellite instability, CD8+ tumor-infiltrating lymphocytes, human leukocyte antigen class I, tumor/specific genotype, and blood biomarkers such as peripheral T-cell phenotype, neutrophil-to-lymphocyte ratio, interferon-gamma, and interleukin-8. A tremendous number of biomarkers are in development, but individual biomarkers are insufficient. Tissue biomarkers have issues in reproducibility and accuracy because of intratumoral heterogeneity and biopsy invasiveness. Furthermore, blood biomarkers have difficulty in reflecting the tumor microenvironment and therefore tend to be less predictive for the efficacy of ICIs than tissue samples. In addition to individual biomarkers, the development of composite markers, including novel technologies such as machine learning and high-throughput analysis, may make it easier to comprehensively analyze multiple biomarkers.
Collapse
Affiliation(s)
| | - Shuji Murakami
- Kanagawa Cancer Center, Department of Thoracic Oncology, 2-3-2 Nakao, Asahi, Yokohama 241-8515, Japan; (R.U.); (H.S.)
| | | |
Collapse
|
24
|
Saleh RR, Scott JL, Meti N, Perlon D, Fazelzad R, Ocana A, Amir E. Prognostic Value of Programmed Death Ligand-1 Expression in Solid Tumors Irrespective of Immunotherapy Exposure: A Systematic Review and Meta-Analysis. Mol Diagn Ther 2022; 26:153-168. [PMID: 35106739 DOI: 10.1007/s40291-022-00576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The programmed cell death-1/programmed cell death ligand-1 (PD-L1) pathway, which plays a crucial role in cancer immune surveillance, is the target of several approved immunotherapeutic agents and is used as a predictive biomarker in some solid tumors. However, its use as a prognostic marker (i.e., regardless of therapy used) is not established clearly with available data demonstrating inconsistent prognostic impact of PD-L1 expression in solid tumors. METHODS We conducted a systematic literature search of electronic databases and identified publications exploring the effect of PD-L1 expression on overall survival and/or disease-free survival. Hazard ratios were pooled in a meta-analysis using generic inverse-variance and random-effects modeling. We used the Deeks method to explore subgroup differences based on disease site, stage of disease, and method of PD-L1 quantification. RESULTS One hundred and eighty-six studies met the inclusion criteria. Programmed cell death ligand-1 expression was associated with worse overall survival (hazard ratio 1.33, 95% confidence interval 1.26-1.39; p < 0.001). There was significant heterogeneity between disease sites (subgroup p = 0.002) with pancreatic, hepatocellular, and genitourinary cancers associated with the highest magnitude of adverse outcomes. Programmed cell death ligand-1 was also associated with worse overall disease-free survival (hazard ratio 1.19, 95% confidence interval 1.09-1.30; p < 0.001). Stage of disease did not significantly affect the results (subgroup p = 0.52), nor did the method of quantification via immunohistochemistry or messenger RNA (subgroup p = 0.70). CONCLUSIONS High expression of PD-L1 is associated with worse survival in solid tumors albeit with significant heterogeneity among tumor types. The effect is consistent in early-stage and metastatic disease and is not sensitive to method of PD-L1 quantification. These data can provide additional information for the counseling of patients with cancer about prognosis.
Collapse
Affiliation(s)
- Ramy R Saleh
- Department of Medical Oncology, McGill University, Montreal, QC, Canada
| | - Jordan L Scott
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Nicholas Meti
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Danielle Perlon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Rouhi Fazelzad
- Information Specialist, Library and Information Services, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Alberto Ocana
- Hospital Clinico San Carlos and Instituto de Investigación Sanitaria San Carlos (IdISSC), and Centro Regional de Investigaciones Biomedicas (CRIB), Centro de Investigación Biomédica en Red Cáncerci (CIBERONC), Universidad Castilla La Mancha (UCLM), Madrid, Spain
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
25
|
Albrecht T, Goeppert B, Brinkmann F, Charbel A, Zhang Q, Schreck J, Wilhelm N, Singer S, Köhler BC, Springfeld C, Mehrabi A, Schirmacher P, Kühl AA, Vogel MN, Jansen H, Utku N, Roessler S. The Transmembrane Receptor TIRC7 Identifies a Distinct Subset of Immune Cells with Prognostic Implications in Cholangiocarcinoma. Cancers (Basel) 2021; 13:cancers13246272. [PMID: 34944891 PMCID: PMC8699724 DOI: 10.3390/cancers13246272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous malignancy with a dismal prognosis. Therapeutic options are largely limited to surgery and conventional chemotherapy offers limited benefit. As immunotherapy has proven highly effective in various cancer types, we have undertaken a quantitative immunohistopathological assessment of immune cells expressing the immunoinhibitory T cell immune response cDNA 7 receptor (TIRC7), an emerging immunoinhibitory receptor, in a cohort of 135 CCA patients. TIRC7+ immune cells were present in both the tumor epithelia and stroma in the majority of CCA cases with the highest levels found in intrahepatic CCA. While intraepithelial density of TIRC7+ immune cells was decreased compared to matched non-neoplastic bile ducts, stromal quantity was higher in the tumor samples. Tumors exhibiting signet ring cell or adenosquamous morphology were exclusively associated with an intraepithelial TIRC7+ phenotype. Survival analysis showed intraepithelial TIRC7+ immune cell density to be a highly significant favorable prognosticator in intrahepatic but not proximal or distal CCA. Furthermore, intraepithelial TIRC7+ immune cell density correlated with the number of intraepithelial CD8+ immune cells and with the total number of CD4+ immune cells. Our results suggest the presence and prognostic relevance of TIRC7+ immune cells in CCA and warrant further functional studies on its pharmacological modulation.
Collapse
Affiliation(s)
- Thomas Albrecht
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Benjamin Goeppert
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Fritz Brinkmann
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Alphonse Charbel
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Qiangnu Zhang
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Johannes Schreck
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Nina Wilhelm
- Tissue Bank of the National Center for Tumor Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Stephan Singer
- Institute of Pathology and Neuropathology, Eberhard-Karls University, 72076 Tübingen, Germany;
| | - Bruno C. Köhler
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Christoph Springfeld
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Arianeb Mehrabi
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
| | - Anja A. Kühl
- Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, iPATH.Berlin, 12203 Berlin, Germany;
| | - Monika N. Vogel
- Diagnostic and Interventional Radiology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany;
| | - Holger Jansen
- Institute for Medical Immunology, Campus Virchow, Charité, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Nalân Utku
- Institute for Medical Immunology, Campus Virchow, Charité, Augustenburger Platz 1, 13353 Berlin, Germany;
- Correspondence: (N.U.); (S.R.); Tel.: +49-23197426350 (N.U.); +49-62215635109 (S.R.)
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; (T.A.); (B.G.); (F.B.); (A.C.); (Q.Z.); (J.S.); (P.S.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (B.C.K.); (C.S.); (A.M.)
- Correspondence: (N.U.); (S.R.); Tel.: +49-23197426350 (N.U.); +49-62215635109 (S.R.)
| |
Collapse
|
26
|
Maniyar RR, Chakraborty S, Jarboe T, Suriano R, Wallack M, Geliebter J, Tiwari RK. Interacting Genetic Lesions of Melanoma in the Tumor Microenvironment: Defining a Viable Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:123-143. [PMID: 34888847 DOI: 10.1007/978-3-030-83282-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Melanoma is the most aggressive form of skin cancer with an estimated 106,110 newly diagnosed cases in the United States of America in 2021 leading to an approximated 7180 melanoma-induced deaths. Cancer typically arises from an accumulation of somatic mutations and can be associated with mutagenic or carcinogenic exposure. A key characteristic of melanoma is the extensive somatic mutation rate of 16.8 mutations/Mb, which is largely attributed to UV exposure. Bearing the highest mutational load, many of them occur in key driver pathways, most commonly the BRAFV600E in the mitogen-activated protein kinase (MAPK) pathway. This driver mutation is targeted clinically with FDA-approved therapies using small molecule inhibitors of oncogenic BRAFV600E and MEK, which has greatly expanded therapeutic intervention following a melanoma diagnosis. Up until 2011, therapeutic options for metastatic melanoma were limited, and treatment typically fell under the spectrum of surgery, radiotherapy, and chemotherapy.Attributed to the extensive mutation rate, as well as having the highest number of neoepitopes, melanoma is deemed to be extremely immunogenic. However, despite this highly immunogenic nature, melanoma is notorious for inducing an immunosuppressive microenvironment which can be relieved by checkpoint inhibitor therapy. The two molecules currently approved clinically are ipilimumab and nivolumab, which target the molecules CTLA-4 and PD-1, respectively.A plethora of immunomodulatory molecules exist, many with redundant functions. Additionally, these molecules are expressed not only by immune cells but also by tumor cells within the tumor microenvironment. Tumor profiling of these cell surface checkpoint molecules is necessary to optimize a clinical response. The presence of immunomodulatory molecules in melanoma, using data from The Cancer Genome Atlas and validation of expression in two model systems, human melanoma tissues and patient-derived melanoma cells, revealed that the expression levels of B and T lymphocyte attenuator (BTLA), TIM1, and CD226, concurrently with the BRAFV600E mutation status, significantly dictated overall survival in melanoma patients. These molecules, along with herpesvirus entry mediator (HVEM) and CD160, two molecules that are a part of the HVEM/BTLA/CD160 axis, had a higher expression in human melanoma tissues when compared to normal skin melanocytes and have unique roles to play in T cell activation. New links are being uncovered between the expression of immunomodulatory molecules and the BRAFV600E genetic lesion in melanoma. Small molecule inhibitors of the MAPK pathway regulate the surface expression of this multifaceted molecule, making BTLA a promising target for immuno-oncology to be targeted in combination with small molecule inhibitors, potentially alleviating T regulatory cell activation and improving patient prognosis.
Collapse
Affiliation(s)
- R R Maniyar
- Human Oncology and Pathogenesis Program, Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Chakraborty
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - T Jarboe
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - R Suriano
- Division of Natural Sciences, College of Mount Saint Vincent, Bronx, NY, USA
| | - M Wallack
- Department Surgery, Metropolitan Hospital, New York, NY, USA
| | - J Geliebter
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - R K Tiwari
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
27
|
Proteomic Implications of Tumoral Infiltrating Lymphocytes in Melanoma: PD-L1, CD4 and CD8 - Short Review. ARS MEDICA TOMITANA 2021. [DOI: 10.2478/arsm-2020-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Melanoma is a tumor developed by malignancy of melanocytes, being one of the most lethal cancers. Usually, it is associated with exposure to ultraviolet radiations, being most common in the skin, but can also be located extracutaneously as in the digestive tract, leptomeninges or uvea. Histopathologically it presents a phase of radial growth and a vertical one, often accompanied by an intra and peritumoral inflammatory infiltrate. Immunohistochemically, the confirmation of the diagnosis of melanoma should be accompanied by the assessment of proteomic markers of lymphocytic infiltrate such as PD-L1, CD4 and CD8. Those have a role in evaluating the prognosis and a possible prediction of the immunotherapeutic response.
Collapse
|
28
|
Affolter A, Lammert A, Kern J, Scherl C, Rotter N. Precision Medicine Gains Momentum: Novel 3D Models and Stem Cell-Based Approaches in Head and Neck Cancer. Front Cell Dev Biol 2021; 9:666515. [PMID: 34307351 PMCID: PMC8296983 DOI: 10.3389/fcell.2021.666515] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the current progress in the development of new concepts of precision medicine for head and neck squamous cell carcinoma (HNSCC), in particular targeted therapies and immune checkpoint inhibition (CPI), overall survival rates have not improved during the last decades. This is, on the one hand, caused by the fact that a significant number of patients presents with late stage disease at the time of diagnosis, on the other hand HNSCC frequently develop therapeutic resistance. Distinct intratumoral and intertumoral heterogeneity is one of the strongest features in HNSCC and has hindered both the identification of specific biomarkers and the establishment of targeted therapies for this disease so far. To date, there is a paucity of reliable preclinical models, particularly those that can predict responses to immune CPI, as these models require an intact tumor microenvironment (TME). The "ideal" preclinical cancer model is supposed to take both the TME as well as tumor heterogeneity into account. Although HNSCC patients are frequently studied in clinical trials, there is a lack of reliable prognostic biomarkers allowing a better stratification of individuals who might benefit from new concepts of targeted or immunotherapeutic strategies. Emerging evidence indicates that cancer stem cells (CSCs) are highly tumorigenic. Through the process of stemness, epithelial cells acquire an invasive phenotype contributing to metastasis and recurrence. Specific markers for CSC such as CD133 and CD44 expression and ALDH activity help to identify CSC in HNSCC. For the majority of patients, allocation of treatment regimens is simply based on histological diagnosis and on tumor location and disease staging (clinical risk assessments) rather than on specific or individual tumor biology. Hence there is an urgent need for tools to stratify HNSCC patients and pave the way for personalized therapeutic options. This work reviews the current literature on novel approaches in implementing three-dimensional (3D) HNSCC in vitro and in vivo tumor models in the clinical daily routine. Stem-cell based assays will be particularly discussed. Those models are highly anticipated to serve as a preclinical prediction platform for the evaluation of stable biomarkers and for therapeutic efficacy testing.
Collapse
Affiliation(s)
- Annette Affolter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | |
Collapse
|
29
|
Ghebeh H, Mansour FA, Colak D, Alfuraydi AA, Al-Thubiti AA, Monies D, Al-Alwan M, Al-Tweigeri T, Tulbah A. Higher PD-L1 Immunohistochemical Detection Signal in Frozen Compared to Matched Paraffin-Embedded Formalin-Fixed Tissues. Antibodies (Basel) 2021; 10:antib10030024. [PMID: 34206205 PMCID: PMC8293136 DOI: 10.3390/antib10030024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/01/2021] [Accepted: 06/18/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose: Response to anti-PD-L1/PD-1 immunotherapy correlates with PD-L1 expression in breast cancer. However, the prevalence of PD-L1 positive breast cancer is variable, which could be due to differences in the population/cohort of patients tested or the preservation/detection technology used. To investigate this variability, we examined the effect of two tissue preservation methods on PD-L1 immunohistochemical detection in breast cancer. Methods: We compared PD-L1 expression in patient-matched frozen (FR) and formalin-fixed paraffin-embedded (FFPE) tissues of breast cancer patients. PD-L1 expression was assessed using tumor proportion score (TPS, simply PD-L1 score), and case positivity was determined with PD-L1 score ≥5. Results: In FFPE tissues, PD-L1 was positive in 7–10% of tested patients, depending on the antibody used. In patient-matched FR tissues, the same antibodies showed positive PD-L1 expression in 20–30% of cases. The impact of the antibody tested on the rate of PD-L1 positivity (% of PDL1 positive cases) was minor, as evident in the near perfect concordance between PD-L1 score obtained using the different antibodies whether tested in FR or FFPE tissues. However, there was a systematic drop by an average of 13–20% in the PD-L1 score obtained in FFPE tissues compared to their patient-matched FR tissues. Conclusions: In the tested patient-matched cohort, there was consistently a higher PD-L1 score in FR than FFPE tissues, regardless of the antibody used, demonstrating a significant effect on PD-L1 detection due to the preservation method. These findings should inspire further work to improve the sensitivity of PD-L1 detection and possibly search for more sensitive antibodies in FFPE tissues.
Collapse
Affiliation(s)
- Hazem Ghebeh
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (F.A.M.); (A.A.A.); (A.A.A.-T.); (M.A.-A.)
- College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
- Correspondence: ; Tel.: +966-1-4424552; Fax: +966-1-4427858
| | - Fatmah A. Mansour
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (F.A.M.); (A.A.A.); (A.A.A.-T.); (M.A.-A.)
| | - Dilek Colak
- Department of Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Akram A. Alfuraydi
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (F.A.M.); (A.A.A.); (A.A.A.-T.); (M.A.-A.)
| | - Amal A. Al-Thubiti
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (F.A.M.); (A.A.A.); (A.A.A.-T.); (M.A.-A.)
| | - Dorota Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Monther Al-Alwan
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (F.A.M.); (A.A.A.); (A.A.A.-T.); (M.A.-A.)
- College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Taher Al-Tweigeri
- Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Asma Tulbah
- Department of Laboratory Medicine and Pathology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| |
Collapse
|
30
|
Longitudinal assessment of PD-L1 expression and gene expression profiles in patients with head and neck cancer reveals temporal heterogeneity. Oral Oncol 2021; 119:105368. [PMID: 34111704 DOI: 10.1016/j.oraloncology.2021.105368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/29/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) is the most validated predictive biomarker used for the treatment of head and neck squamous cell carcinoma (HNSCC) with immune checkpoint inhibitors (ICI). Several gene expression-based signatures surrogate of the activation of IFN-gamma pathway and of the presence of tertiary lymphoid structures (TLS) have also been proposed as potential biomarkers. While they may have a potential therapeutic implication, the longitudinal changes of either PD-L1 or gene expression profiles between the initial and recurrent HNSCC lesions is unknown. METHODS PD-L1 immunohistochemistry (IHC) and targeted RNA-sequencing of 2,549 transcripts were analyzed on paired specimens from the initial diagnosis and recurrent HNSCC. PD-L1 status was defined using the combined positive score (CPS). PD-L1 mRNA levels were compared with protein expression levels by IHC. Enrichment scores of surrogate signatures for TLS and IFN-gamma (IFN-γ) pathway activation were computed using the single sample gene set enrichment analysis (ssGSEA). RESULTS PD-L1 status was 64% (21/33) concordant between the initial and recurrent lesions using a CPS 1 threshold and 67% (22/33) concordant using a CPS 20 threshold. CPS score was associated with PD-L1 gene expression levels. There was a 43% (15/35) and 66% (23/35) concordance for the IFN-γ and TLS signature scores, respectively. CONCLUSION Our study reveals temporal heterogeneity of PD-L1 status and TLS/IFN-γ gene expression surrogates in HNSCC that need to be considered when interpreting biomarker studies.
Collapse
|
31
|
Yazdanpanah P, Alavianmehr A, Ghaderi A, Monabati A, Montazer M, Tahmasbi K, Farjadian S. PD-L1 expression in tumor lesions and soluble PD-L1 serum levels in patients with breast cancer: TNBC versus TPBC. Breast Dis 2021; 40:43-50. [PMID: 33523034 DOI: 10.3233/bd-201049] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Block of programmed cell death protein 1 (PD-1) interaction with its ligand, PD-L1, enhances anti-tumor activity. OBJECTIVES We aimed to assess the association between PD-L1 expression in tumor cells and CD8+ tumor infiltrating T cells (TILs) as well as soluble (s)PD-L1 serum levels in patients with triple negative breast cancer (TNBC) compared to triple positive (TPBC). METHODS A total of 113 tumor sections and 133 serum samples were available from 144 patients with breast cancer (72 TNBC and 72 TPBC). Dual immunohistochemistry staining was applied to determine differential PD-L1 expression in tumor cells and CD8+ TILs. Soluble PD-L1 serum levels were also evaluated in patients compared to 40 healthy women by ELISA method. RESULTS Despite TPBC patients which were mostly grades 1/2, TNBC patients were grade 3 (72% versus 66.7%, P < 0.001). Most of the TNBC patients were stages I/II, whereas most of the TPBC patients were stages III/IV (57.3% versus 68.3%,P = 0.005). There was no difference in tumor size and metastasis between TNBC and TPBC patients, although the number of involved lymph nodes was significantly more in TPBC patients (P = 0.0012). PD-L1 expression was detected in 11.5% of samples mostly in TNBC subtype and was associated with advanced grades (P = 0.039). There was no relationship between PD-L1 expression and tumor stage. PD-L1 expression in CD8+ TILs was nonsignificantly higher than tumor cells. Serum levels of sPD-L1 showed no difference between patients and healthy women. We found no correlation between PD-L1 expression in tumor lesions and serum levels of sPD-L1 in patients. CONCLUSION PD-L1 expression was more detected in our patients with TNBC. It seems that, these patients who are resistant to standard chemotherapy regimens may get benefit from PD-L1 inhibition therapy and because of its low serum levels, sPD-L1 cannot interfere with this therapy.
Collapse
Affiliation(s)
| | - Ali Alavianmehr
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran.,Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Monabati
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Montazer
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamran Tahmasbi
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Farjadian
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Guo D, Ji X, Luo J. Rational nanocarrier design towards clinical translation of cancer nanotherapy. Biomed Mater 2021; 16. [DOI: 10.1088/1748-605x/abe35a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
|
33
|
Qiu Z, Du Y. Clinicopathological and prognostic significance of programmed death ligant-1 expression in gastric cancer: a meta-analysis. J Gastrointest Oncol 2021; 12:112-120. [PMID: 33708429 DOI: 10.21037/jgo-20-568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Gastric cancer (GC) is a common malignant tumor with a high incidence in China. The use of immune checkpoint inhibitors has become the focus of tumor immunotherapy in recent years. This study was to investigate the clinicopathological and prognostic significance of programmed death ligant-1 (PD-L1) expression in GC. Methods We searched the PubMed, ScienceNet, EMbase, CNKI, and Wanfang databases for retrospective cohort studies on the clinicopathology and prognosis of PD-L1 expression in GC published between January 2010 and April 2020. The literature was first selected to extract data according to the inclusion and exclusion criteria, then a meta-analysis performed using Stata15.0 software. Publication bias and sensitivity analysis were carried out for the included studies. Results A total of 3,218 patients in 15 studies were included in the meta-analysis. The positive expression of PD-L1 was related to a decrease in the 3-year survival rate (HR =1.32, 95% CI: 1.02-1.49, P=0.028) and 5-year survival rate (HR =1.39, 95% CI: 1.14-1.69, P=0.001). The difference in PD-L1 expression was related to lymph node metastasis (OR =1.73, 95% CI: 1.18-2.54, P<0.001), but not to tumor stage (OR =1.28, 95% CI: 0.81-2.02, P=0.292). Conclusions The results show that PD-L1 is related to the prognosis of GC. Its high expression decreases the 3- and 5-year survival rates and promotes lymph node metastasis, but does not reflect tumor stage. The results may provide a theoretical basis for the choice of clinical immunotherapy in GC patients.
Collapse
Affiliation(s)
- Zhebing Qiu
- Department of Gastrointestinal Tumor Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, China
| | - Yinguo Du
- Department of Gastrointestinal Tumor Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, China
| |
Collapse
|
34
|
Trailblazing perspectives on targeting breast cancer stem cells. Pharmacol Ther 2021; 223:107800. [PMID: 33421449 DOI: 10.1016/j.pharmthera.2021.107800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer (BCa) is one of the most prevalent malignant tumors affecting women's health worldwide. The recurrence and metastasis of BCa have made it a long-standing challenge to achieve remission-persistent or disease-undetectable clinical outcomes. Cancer stem cells (CSCs) possess the ability to self-renew and generate heterogeneous tumor bulk. The existence of CSCs has been found to be vital in the initiation, metastasis, therapy resistance, and recurrence of tumors across cancer types. Because CSCs grow slowly in their dormant state, they are insensitive to conventional chemotherapies; however, when CSCs emerge from their dormant state and become clinically evident, they usually acquire genetic traits that make them resistant to existing therapies. Moreover, CSCs also show evidence of acquired drug resistance in synchrony with tumor relapses. The concept of CSCs provides a new treatment strategy for BCa. In this review, we highlight the recent advances in research on breast CSCs and their association with epithelial-mesenchymal transition (EMT), circulating tumor cells (CTCs), plasticity of tumor cells, tumor microenvironment (TME), T-cell modulatory protein PD-L1, and non-coding RNAs. On the basis that CSCs are associated with multiple dysregulated biological processes, we envisage that increased understanding of disease sub-classification, selected combination of conventional treatment, molecular aberration directed therapy, immunotherapy, and CSC targeting/sensitizing strategy might improve the treatment outcome of patients with advanced BCa. We also discuss novel perspectives on new drugs and therapeutics purposing the potent and selective expunging of CSCs.
Collapse
|
35
|
The application of nano-medicine to overcome the challenges related to immune checkpoint blockades in cancer immunotherapy: Recent advances and opportunities. Crit Rev Oncol Hematol 2021; 157:103160. [DOI: 10.1016/j.critrevonc.2020.103160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/27/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
|
36
|
Bahreyni A, Mohamud Y, Luo H. Emerging nanomedicines for effective breast cancer immunotherapy. J Nanobiotechnology 2020; 18:180. [PMID: 33298099 PMCID: PMC7727246 DOI: 10.1186/s12951-020-00741-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer continues to be the most frequently diagnosed malignancy among women, putting their life in jeopardy. Cancer immunotherapy is a novel approach with the ability to boost the host immune system to recognize and eradicate cancer cells with high selectivity. As a promising treatment, immunotherapy can not only eliminate the primary tumors, but also be proven to be effective in impeding metastasis and recurrence. However, the clinical application of cancer immunotherapy has faced some limitations including generating weak immune responses due to inadequate delivery of immunostimulants to the immune cells as well as uncontrolled modulation of immune system, which can give rise to autoimmunity and nonspecific inflammation. Growing evidence has suggested that nanotechnology may meet the needs of current cancer immunotherapy. Advanced biomaterials such as nanoparticles afford a unique opportunity to maximize the efficiency of immunotherapy and significantly diminish their toxic side-effects. Here we discuss recent advancements that have been made in nanoparticle-involving breast cancer immunotherapy, varying from direct activation of immune systems through the delivery of tumor antigens and adjuvants to immune cells to altering immunosuppression of tumor environment and combination with other conventional therapies.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard St, Vancouver, BC, V6Z 1Y6, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard St, Vancouver, BC, V6Z 1Y6, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard St, Vancouver, BC, V6Z 1Y6, Canada. .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
37
|
Yang B, Gao J, Pei Q, Xu H, Yu H. Engineering Prodrug Nanomedicine for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002365. [PMID: 33304763 PMCID: PMC7709995 DOI: 10.1002/advs.202002365] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/16/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy has shifted the clinical paradigm of cancer management. However, despite promising initial progress, immunotherapeutic approaches to cancer still suffer from relatively low response rates and the possibility of severe side effects, likely due to the low inherent immunogenicity of tumor cells, the immunosuppressive tumor microenvironment, and significant inter- and intratumoral heterogeneity. Recently, nanoformulations of prodrugs have been explored as a means to enhance cancer immunotherapy by simultaneously eliciting antitumor immune responses and reversing local immunosuppression. Prodrug nanomedicines, which integrate engineering advances in chemistry, oncoimmunology, and material science, are rationally designed through chemically modifying small molecule drugs, peptides, or antibodies to yield increased bioavailability and spatiotemporal control of drug release and activation at the target sites. Such strategies can help reduce adverse effects and enable codelivery of multiple immune modulators to yield synergistic cancer immunotherapy. In this review article, recent advances and translational challenges facing prodrug nanomedicines for cancer immunotherapy are overviewed. Last, key considerations are outlined for future efforts to advance prodrug nanomedicines aimed to improve antitumor immune responses and combat immune tolerogenic microenvironments.
Collapse
Affiliation(s)
- Bin Yang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Qing Pei
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Huixiong Xu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
38
|
Sui X, Jin T, Liu T, Wu S, Wu Y, Tang Z, Ren Y, Ni D, Yao Z, Zhang H. Tumor Immune Microenvironments (TIMEs): Responsive Nanoplatforms for Antitumor Immunotherapy. Front Chem 2020; 8:804. [PMID: 33094098 PMCID: PMC7508192 DOI: 10.3389/fchem.2020.00804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022] Open
Abstract
Interest in cancer immunotherapy has rapidly risen since it offers many advantages over traditional approaches, such as high efficiency and prevention of metastasis. Efforts have primarily focused on two major strategies for regulating the body's antitumor immune response mechanisms: “enhanced immunotherapy” that aims to amplify the immune activation, and “normalized immunotherapy” that corrects the defective immune mechanism in the tumor immune microenvironments (TIMEs), which returns to the normal immune trajectory. However, due to the complexity and heterogeneity of the TIMEs, and lack of visualization research on the immunotherapy process, cancer immunotherapy has not been widely used in clinical setting. Recently, through the design and modification of nanomaterials, intelligent TIME-responsive nanoplatforms were developed from which encouraging results in many aspects of immunotherapy have been achieved. In this mini review, the status of designed nanomaterials for nanoplatform-based immune regulation of TIMEs has been emphasized, particularly with respect to the aforementioned approaches. It is envisaged that future prospects will focus on a combination of multiple immunotherapies for more efficient cancer inhibition and elimination.
Collapse
Affiliation(s)
- Xueqing Sui
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Teng Jin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tonghui Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiman Wu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yue Wu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongmin Tang
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Yan Ren
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dalong Ni
- Departments of Radiology, Medical Physics, and Pharmaceutical Sciences, University of Wisconsin - Madison, Madison, WI, United States
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hua Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
39
|
Gupta S, Zugazagoitia J, Martinez-Morilla S, Fuhrman K, Rimm DL. Digital quantitative assessment of PD-L1 using digital spatial profiling. J Transl Med 2020; 100:1311-1317. [PMID: 32249818 PMCID: PMC7502436 DOI: 10.1038/s41374-020-0424-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 12/04/2022] Open
Abstract
The assessment of programmed death 1 ligand 1 (PD-L1) expression by Immunohistochemistry (IHC) is the US Food and Drug Administration (FDA)-approved predictive marker to select responders to checkpoint blockade anti-PD-1/PD-L1 axis immunotherapies. Different PD-L1 immunohistochemistry (IHC) assays use different antibodies and different scoring methods in tumor cells and immune cells. Multiple studies have compared the performance of these assays with variable results. Here, we investigate an alternative method for assessment of PD-L1 using a new technology known as digital spatial profiling. We use a previously described standardization tissue microarray (TMA) to assess the accuracy of the method and compare digital spatial profiler (DSP) to each FDA-approved PD-L1 assays, one LDT assay and three quantitative fluorescence assays. The standardized cell line Index tissue microarray contains 10 isogenic cells lines in triplicates expressing various ranges of PD-L1. The dynamic range of PD-L1 digital counts was measured in the ten cell lines on the Index TMA using the GeoMx DSP assay and read on the nCounter platform. The digital method shows very high correlation with immunohistochemistry scored with quantitative software and with quantitative fluorescence. High correlation of PD-L1 digital DSP counts were seen between rows on the same Index TMA. Finally, experiments from two Index TMAs showed reproducibility of DSP counts were independent of variable slide storage time over a three-week period after antibody labeling but before collection of cleaved tags. In summary, DSP appears to have quantitative potential comparable to quantitative immunohistochemistry. It is possible that this technology could be used as a PD-L1 protein measurement system for companion diagnostic testing for immune therapy.
Collapse
Affiliation(s)
- Swati Gupta
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jon Zugazagoitia
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
40
|
Caldaria A, Giuffrida R, di Meo N, Massari L, Dianzani C, Cannavò SP, Degrassi F, Casablanca E, Zalaudek I, Conforti C. Diagnosis and treatment of melanoma bone metastasis: A multidisciplinary approach. Dermatol Ther 2020; 33:e14193. [DOI: 10.1111/dth.14193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Antonio Caldaria
- Orthopedic and Traumatology Unit Sant'Anna University Hospital Ferrara Italy
| | - Roberta Giuffrida
- Department of Clinical and Experimental Medicine, Dermatology University of Messina Messina Italy
| | - Nicola di Meo
- Dermatology Clinic, Hospital Maggiore of Trieste University of Trieste Trieste Italy
| | - Leo Massari
- Orthopedic and Traumatology Unit Sant'Anna University Hospital Ferrara Italy
| | - Caterina Dianzani
- Plastic Surgery Unit, Section of Dermatology Campus Biomedico University Rome Italy
| | | | - Ferruccio Degrassi
- Department of Radiology, Maggiore Hospital of Trieste University of Trieste Trieste Italy
| | - Edoardo Casablanca
- Department of Orthopedics and Trauma Surgery University of Verona Verona Italy
| | - Iris Zalaudek
- Dermatology Clinic, Hospital Maggiore of Trieste University of Trieste Trieste Italy
| | - Claudio Conforti
- Dermatology Clinic, Hospital Maggiore of Trieste University of Trieste Trieste Italy
| |
Collapse
|
41
|
Hu Z, Lu L, Fei Z, Lv D. Association between clinicopathological features and prognosis significance of PD-L1 expression in small cell lung cancer patients: a systemic review and meta-analysis. Transl Cancer Res 2020; 9:5508-5516. [PMID: 35117915 PMCID: PMC8798485 DOI: 10.21037/tcr-20-1512a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Programmed death ligand-1 (PD-L1) has been identified as an established biomarker for predicting response to immunotherapy in a variety types of cancer. However, the clinicopathological and prognostic significance of this protein in small cell lung cancer (SCLC) patients remains controversial. METHODS Eligible studies extracted from the databases of PubMed, MEDLINE, Embase, and CNKI databases were evaluated. Statistical analysis was performed using STATA 11.2 software. RESULTS A total of 483 PD-L1+ cases and 570 controls from 11 publications were extracted. Either overall analysis or subcategory analysis showed that no significant association between higher PD-L1 expression and gender (n=8, OR 1.08, 95% CI: 0.73-1.61, P=0.704, I2=0.0%), tumor stage (n=5, OR 0.71, 95% CI: 0.20-2.56, P=0.599, I2=86.5%), smoking status (n=4, OR 0.85, 95% CI: 0.41-1.73, P=0.646, I2=0.0%), and the level of serum lactate dehydrogenase (LDH) (n=4, OR 0.76, 95% CI: 0.48-1.20, P=0.241, I2= 21.6%). PD-L1 expression had no positive correlation with overall survival (OS) (n=11, HR 0.97, 95% CI: 0.61-1.56, P=0.904, I2= 83.2%) in overall analysis. However, the stratified analysis showed that increased expression of PD-L1 predicted a significantly better OS in monoclonal antibody (mAb) subgroup and Food and Drug Administration (FDA) approved antibody clone specification (22C3/28-8/SP142/SP263) subgroup without significant heterogeneity. CONCLUSIONS PD-L1 is not an important predictor of most clinicopathological features of SCLC patients, but it can predict an improved survival when using mAb or FDA approved clone specifications in IHC assays.
Collapse
Affiliation(s)
- Zongtao Hu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Lin Lu
- Department of Clinical Oncology, 901 Hospital of Joint Logistics Support Force of People Liberation Army, Hefei, China
| | - Zhenle Fei
- Department of Clinical Oncology, 901 Hospital of Joint Logistics Support Force of People Liberation Army, Hefei, China
| | - Donglai Lv
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China.,Department of Clinical Oncology, 901 Hospital of Joint Logistics Support Force of People Liberation Army, Hefei, China
| |
Collapse
|
42
|
Ma H, Wang H, Sove RJ, Jafarnejad M, Tsai CH, Wang J, Giragossian C, Popel AS. A Quantitative Systems Pharmacology Model of T Cell Engager Applied to Solid Tumor. AAPS JOURNAL 2020; 22:85. [PMID: 32533270 PMCID: PMC7293198 DOI: 10.1208/s12248-020-00450-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/20/2020] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has recently drawn remarkable attention as promising results in the clinic have shown its ability to improve the overall survival, and T cells are considered to be one of the primary effectors for cancer immunotherapy. Enhanced and restored T cell tumoricidal activity has shown great potential for killing cancer cells. Bispecific T cell engagers (TCEs) are a growing class of molecules that are designed to bind two different antigens on the surface of T cells and cancer cells to bring them in close proximity and selectively activate effector T cells to kill target cancer cells. New T cell engagers are being investigated for the treatment of solid tumors. The activity of newly developed T cell engagers showed a strong correlation with tumor target antigen expression. However, the correlation between tumor-associated antigen expression and overall response of cancer patients is poorly understood. In this study, we used a well-calibrated quantitative systems pharmacology (QSP) model extended to bispecific T cell engagers to explore their efficacy and identify potential biomarkers. In principle, patient-specific response can be predicted through this model according to each patient's individual characteristics. This extended QSP model has been calibrated with available experimental data and provides predictions of patients' response to TCE treatment.
Collapse
Affiliation(s)
- Huilin Ma
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard J Sove
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chia-Hung Tsai
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Jun Wang
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Craig Giragossian
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Soluble programmed death-ligand 1 rather than PD-L1 on tumor cells effectively predicts metastasis and prognosis in soft tissue sarcomas. Sci Rep 2020; 10:9077. [PMID: 32493964 PMCID: PMC7270095 DOI: 10.1038/s41598-020-65895-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
The soluble form of PD-L1 (sPD-L1) is related to a poor prognosis in various cancers. Comparisons of sPD-L1 and PD-L1 expressed on tumor cells in soft tissue tumor patients have not been reported. The purpose of this study was to analyze serum sPD-L1 and PD-L1 levels in soft tissue tumor patients. A total of 135 patients with primary soft tissue tumors were enrolled in this study. The sPD-L1 level was quantitatively measured by enzyme immunoassay, and PD-L1 expression on high grade sarcoma cells was analyzed immunohistologically. There were no significant differences in sPD-L1 levels between benign (48) and soft tissue sarcoma (STS) patients (87). In STS, the high sPD-L1 (>44.26 pg/mL) group had significantly lower metastasis-free survival (MS) and lower overall survival (OS) than the low sPD-L1 group (≤44.26 pg/mL) at 5 years using the log-rank test. On multivariate Cox proportional hazard analysis, the high sPD-L1 group had significant differences in MS and OS compared to the low sPD-L1 group. Between positive and negative immunostaining groups, recurrence-free survival (RS), MS, and OS were not significantly different. No correlation was found between immunostaining and sPD-L1 with the Kappa coefficient. The sPD-L1 concentration could predict future metastasis and prognosis in STS patients. High sPD-L1 in STS patients may be a target for treatment with checkpoint inhibitors.
Collapse
|
44
|
PD-1, PD-L1, and BIM as Predictors of Sentinel Lymph Node Metastasis in Primary Cutaneous Melanoma. J Invest Dermatol 2020; 140:2301-2304.e3. [PMID: 32275976 DOI: 10.1016/j.jid.2020.03.948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 11/20/2022]
|
45
|
Xu J, Wang F, Yan Y, Zhang Y, Du Y, Sun G. Prognostic and Clinicopathological Value of PD-L1 in Melanoma: A Meta-Analysis. Am J Med Sci 2020; 359:339-346. [PMID: 32498941 DOI: 10.1016/j.amjms.2020.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/08/2020] [Accepted: 03/25/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND There is a growing interest in using programmed death ligand-1 (PD-L1) as a prognostic marker for melanoma. We conducted this meta-analysis to explore the prognostic and clinicopathological value of PD-L1 in melanoma. MATERIALS AND METHODS The electronic databases PubMed, Web of Science and the Cochrane Library were searched for relevant studies. The major investigated parameters were PD-L1 expression levels in relation to patient gender, tumor-infiltrating lymphocytes (TILs), tumor stage, lymph node (LN) metastasis, histological type, progression-free survival (PFS) and overall survival (OS). Odds ratios (ORs) and hazard ratios (HRs) were computed using the fixed-effect or random-effects model according to data heterogeneity. RESULTS Positive PD-L1 expression was significantly associated with high levels of TILs (OR = 7.56, 95% CI 2.04-28.02), metastatic melanoma (OR = 0.45, 95% CI 0.30-0.67) and LN-positive melanoma (OR = 2.56, 95% CI 1.31-4.99) but not gender or histological type. In addition, the pooled HRs showed no relation between PD-L1 expression and PFS (HR = 1.18, 95% CI 0.83-1.69) or OS (HR = 0.77, 95% CI 0.47-1.25). When restricted to metastatic melanoma, positive PD-L1 expression was significantly related to prolonged OS (HR = 0.57, 95% CI 0.46-0.70). CONCLUSIONS Positive PD-L1 expression may be an important prognostic factor for longer OS in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Jing Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fang Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunfang Yan
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yiruo Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yingying Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
46
|
Nanoparticle mediated cancer immunotherapy. Semin Cancer Biol 2020; 69:307-324. [PMID: 32259643 DOI: 10.1016/j.semcancer.2020.03.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022]
Abstract
The versatility and nanoscale size have helped nanoparticles (NPs) improve the efficacy of conventional cancer immunotherapy and opened up exciting approaches to combat cancer. This review first outlines the tumor immune evasion and the defensive tumor microenvironment (TME) that hinders the activity of host immune system against tumor. Then, a detailed description on how the NP based strategies have helped improve the efficacy of conventional cancer vaccines and overcome the obstacles led by TME. Sustained and controlled drug delivery, enhanced cross presentation by immune cells, co-encapsulation of adjuvants, inhibition of immune checkpoints and intrinsic adjuvant like properties have aided NPs to improve the therapeutic efficacy of cancer vaccines. Also, NPs have been efficient modulators of TME. In this context, NPs facilitate better penetration of the chemotherapeutic drug by dissolution of the inhibitory meshwork formed by tumor associated cells, blood vessels, soluble mediators and extra cellular matrix in TME. NPs achieve this by suppression, modulation, or reprogramming of the immune cells and other mediators localised in TME. This review further summarizes the applications of NPs used to enhance the efficacy of cancer vaccines and modulate the TME to improve cancer immunotherapy. Finally, the hurdles faced in commercialization and translation to clinic have been discussed and intriguingly, NPs owe great potential to emerge as clinical formulations for cancer immunotherapy in near future.
Collapse
|
47
|
Yang J, Dong M, Shui Y, Zhang Y, Zhang Z, Mi Y, Zuo X, Jiang L, Liu K, Liu Z, Gu X, Shi Y. A pooled analysis of the prognostic value of PD-L1 in melanoma: evidence from 1062 patients. Cancer Cell Int 2020; 20:96. [PMID: 32256205 PMCID: PMC7106672 DOI: 10.1186/s12935-020-01187-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/23/2020] [Indexed: 02/08/2023] Open
Abstract
Background Programmed death-ligand 1 (PD-L1) was the first identified ligand of programmed death-1 (PD-1). PD-1/PD-L1 interactions inhibit T cell-mediated immune responses, limit cytokine production, and promote tumor immune escape. Recently, many studies have investigated the prognostic value of PD-L1 expression in patients with melanoma. However, the results of these analyses remain a subject of debate. We have therefore carried out a meta-analysis to identify the prognostic role of PD-L1 in melanoma. Methods A thorough medical literature search was performed in the databases PubMed, Web of Science, and Embase until October 2019. The pooled hazard ratios (HRs) and 95% confidence intervals (95% CIs) were calculated to evaluate the correlation between PD-L1 overexpression and prognosis. Publication bias was evaluated using Begg’s test and Egger’s test. Results Thirteen articles with 1062 enrolled patients were included in this meta-analysis. High PD-L1 expression did not correlate with overall survival (OS) (HR = 0.93, 95% CI 0.57–1.52, P = 0.781) or progression-free survival (PFS) (HR = 0.82, 95% CI 0.43–1.54, P = 0.535). However, PD-L1 overexpression correlated with the absence of lymph node (LN) metastasis (OR = 0.46, 95% CI 0.22–0.95, P = 0.036). Further, there was no significant relationship between PD-L1 expression and sex (OR = 1.29, 95% CI 0.90–1.84, P = 0.159), age (OR = 0.90, 95% CI 0.51–1.57, P = 0.708), or Eastern Cooperative Oncology Group Performance Status (OR = 0.55, 95% CI 0.06–4.83, P = 0.592). Conclusions This meta-analysis suggested that PD-L1 expression did not predict an inferior prognosis in patients with melanoma. However, high PD-L1 expression was associated with absence of LN metastasis in such patients.
Collapse
Affiliation(s)
- Jing Yang
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Meilian Dong
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Yifang Shui
- 2Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Yue Zhang
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Zhigang Zhang
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Yin Mi
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Xiaoxiao Zuo
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Li Jiang
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Ke Liu
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Zheyan Liu
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Xiaobin Gu
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| | - Yonggang Shi
- 1Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People's Republic of China
| |
Collapse
|
48
|
Shen J, Li S, Medeiros LJ, Lin P, Wang SA, Tang G, Yin CC, You MJ, Khoury JD, Iyer SP, Miranda RN, Xu J. PD-L1 expression is associated with ALK positivity and STAT3 activation, but not outcome in patients with systemic anaplastic large cell lymphoma. Mod Pathol 2020; 33:324-333. [PMID: 31383967 DOI: 10.1038/s41379-019-0336-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022]
Abstract
The programmed cell death 1 (PD-1) pathway is a recently recognized mechanism of tumor immune evasion. In this study, programmed cell death ligand 1 (PD-L1) expression was evaluated in 95 patients with systemic anaplastic large cell lymphoma: 45 ALK+ and 50 ALK-. ALK+ anaplastic large cell lymphoma was more often positive for PD-L1 than ALK- anaplastic large cell lymphoma (76% vs 42%, p = 0.002). ALK- anaplastic large cell lymphoma showed a strong correlation between PD-L1 expression and STAT3 activation (measured by pSTAT3Tyr705) (r = 0.8, p < 0.0001). In contrast, the PD-L1/pSTAT3 correlation was weaker in ALK+ anaplastic large cell lymphoma (r = 0.4, p = 0.08). In ALK- anaplastic large cell lymphoma, the PD-L1+ subgroup was more often EMA positive (69% vs 20%, p = 0.02) and tended to be less often CD2+ (50% vs 83%, p = 0.059). In ALK+ anaplastic large cell lymphoma, PD-L1 was not associated with pathologic features (all p > 0.05). Negative ALK status and high IPI score (≥3) were associated with shorter overall survival (p = 0.009 and p = 0.0005, respectively). Overall survival was not different between patients with PD-L1+ vs PD-L1- anaplastic large cell lymphoma (p = 0.44), regardless of ALK status and International Prognostic Index (IPI) score. We conclude that PD-L1 expression is more common in ALK+ anaplastic large cell lymphoma than ALK- anaplastic large cell lymphoma. In ALK- anaplastic large cell lymphoma, PD-L1 is strongly correlated with STAT3 activation and is associated with more frequent EMA and less frequent CD2 expression. PD-L1 has no prognostic significance in predicting the outcome of patients with systemic anaplastic large cell lymphoma, regardless of ALK status. PD-L1 expression on the anaplastic large cell lymphoma cells suggests these patients as potential candidates for PD-1 blockade immunotherapy.
Collapse
Affiliation(s)
- Jing Shen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Hematology, Capital Medical University Beijing Friendship Hospital, Beijing, China
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C Cameron Yin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph D Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Swaminathan P Iyer
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Xu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
49
|
Lippens L, Van Bockstal M, De Jaeghere EA, Tummers P, Makar A, De Geyter S, Van de Vijver K, Hendrix A, Vandecasteele K, Denys H. Immunologic impact of chemoradiation in cervical cancer and how immune cell infiltration could lead toward personalized treatment. Int J Cancer 2020; 147:554-564. [PMID: 32017078 DOI: 10.1002/ijc.32893] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023]
Abstract
We investigated the potential of tumor-infiltrating immune cells (ICs) as predictive or prognostic biomarkers for cervical cancer patients. In total, 38 patients treated with (chemo)radiotherapy and subsequent surgery were included in the current study. This unique treatment schedule makes it possible to analyze IC markers in pretreatment and posttreatment tissue specimens and their changes during treatment. IC markers for T cells (CD3, CD4, CD8 and FoxP3), macrophages (CD68 and CD163) and B cells (CD20), as well as IL33 and PD-L1, were retrospectively analyzed via immunohistochemistry. Patients were grouped in the low score or high score group based on the amount of positive cells on immunohistochemistry. Correlations to pathological complete response (pCR), cause-specific survival (CSS) and metastasis development during follow-up were evaluated. In analysis of pretreatment biopsies, significantly more pCR was seen for patients with CD8 = CD3, CD8 ≥ CD4, positive IL33 tumor cell (TC) scores, IL33 IC < TC and PD-L1 TC ≥5%. Besides patients with high CD8 scores, also patients with CD8 ≥ CD4, CD163 ≥ CD68 or PD-L1 IC ≥5% had better CSS. In the analysis of posttreatment specimens, less pCR was observed for patients with high CD8 or CD163 scores. Patients with decreasing CD8 or CD163 scores between pretreatment and posttreatment samples showed more pCR, whereas those with increasing CD8 or decreasing IL33 IC scores showed a worse CSS. Meanwhile, patients with an increasing CD3 score or stable/increasing PD-L1 IC score showed more metastasis during follow-up. In this way, the intratumoral IC landscape is a promising tool for prediction of outcome and response to (chemo)radiotherapy.
Collapse
Affiliation(s)
- Lien Lippens
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium.,Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Mieke Van Bockstal
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium.,Pathology, Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Emiel A De Jaeghere
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium.,Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Philippe Tummers
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Gynecology, Department of Human Structure and Repair, Gent University Hospital, Ghent, Belgium
| | - Amin Makar
- Gynecology, Department of Human Structure and Repair, Gent University Hospital, Ghent, Belgium
| | - Sofie De Geyter
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium.,Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Koen Van de Vijver
- Pathology, Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Katrien Vandecasteele
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Radiation Therapy, Department of Human Structure and Repair, Ghent University Hospital, Ghent, Belgium
| | - Hannelore Denys
- Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
50
|
Ren Y, Lv Q, Yue W, Liu B, Zou Z. The programmed cell death protein-1/programmed cell death ligand 1 expression, CD3+ T cell infiltration, NY-ESO-1 expression, and microsatellite instability phenotype in primary cutaneous melanoma and mucosal melanoma and their clinical significance and prognostic value: a study of 89 consecutive cases. Melanoma Res 2020; 30:85-101. [PMID: 31095042 DOI: 10.1097/cmr.0000000000000620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We evaluated the expression of programmed cell death protein-1 (PD-1), programmed cell death ligand 1 (PD-L1), and NY-ESO-1 antigen; the infiltration of CD3+ T cells; and the microsatellite instability (MSI) phenotype, as well as the relationship of each factor to survival in malignant melanoma patients. Malignant melanoma samples from 89 patients were stained by immunohistochemistry to evaluate PD-1, PD-L1, CD3+ tumor-infiltrating lymphocytes (TILs), NY-ESO-1, and MSI. PD-1 and PD-L1 were expressed in 19.1 and 32.6% of the 89 samples, respectively. There was a significant correlation between PD-1 and PD-L1 expression (r = 0.207, P = 0.046). High infiltration of CD3+ T cells was observed in 41.6% of the samples, and increased cell infiltration was associated with increased PD-1 expression (P = 0.001). NY-ESO-1 antigen was detected in 13.5% of all samples, and the expression of NY-ESO-1 was positively correlated with the expression of PD-1 (P < 0.001). In our research, MSI was detected in 18 samples (20.2%). Survival analysis showed that a high infiltration of CD3+ T cells was related to longer progression-free survival (PFS) [24.0 months, 95% confidence interval (CI): 7.4-40.6 vs. 11.0 months, 95% CI: 7.1-12.9, P = 0.031], similarly, the median overall survival (OS) of the CD3+ T cell high-infiltration patients was also longer (53.0 vs. 38.0 months), but with no statistical significance (P = 0.200). The results for the immune markers mentioned above provide a theoretical basis for the prognosis and immunotherapy selection of malignant melanoma patients.
Collapse
Affiliation(s)
- Yu Ren
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
| | - Qing Lv
- Yi Xing Tumor Hospital, Yixing, China
| | - Wuheng Yue
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing
| | - Baorui Liu
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
| | - Zhengyun Zou
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
| |
Collapse
|