1
|
Wiseman CL, Holmes JP, Calfa C, Dakhil SR, Bhattacharya S, Peoples GE, Lacher MD, Lopez-Lago M, Kharazi A, Del Priore G, Chang M, Adams DL, Williams WV. Results of a phase I/IIa trial of SV-BR-1-GM inoculation with low-dose cyclophosphamide and interferon alpha (Bria-IMT) in metastatic breast cancer. Hum Vaccin Immunother 2024; 20:2379864. [PMID: 39165083 PMCID: PMC11340742 DOI: 10.1080/21645515.2024.2379864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 08/22/2024] Open
Abstract
This Phase I/IIa open-label, single-arm clinical trial addressing advanced, refractory, metastatic breast cancer was conducted at six medical centers in the United States. We repeated inoculations with irradiated SV-BR-1-GM, a breast cancer cell line with antigen-presenting activity engineered to release granulocyte-macrophage colony-stimulating factor (GM-CSF), with pre-dose low-dose cyclophosphamide and post-dose local interferon alpha. Twenty-six patients were enrolled; 23 (88.5%) were inoculated, receiving a total of 79 inoculations. There were six Grade 4 and one Grade 5 adverse events noted (judged unrelated to SV-BR-1-GM). Disease control (stable disease [SD]) occurred in 8 of 16 evaluable patients; 4 showed objective regression of metastases, including 1 patient with near-complete regressions in 20 of 20 pulmonary lesions. All patients with regressions had human leukocyte antigen (HLA) matches with SV-BR-1-GM; non-responders were equally divided between matching and nonmatching (p = .01, Chi-squared), and having ≥2 HLA matches with SV-BR-1-GM (n = 6) correlated with clinical benefit. Delayed-type hypersensitivity (DTH) testing to candida antigen and SV-BR-1-GM generated positive responses (≥5 mm) in 11 (42.3%) and 13 (50%) patients, respectively. Quantifying peripheral circulating tumor cells (CTCs) and cancer-associated macrophage-like cells (CAMLs) showed that a drop in CAMLs was significantly correlated with an improvement in progression-free survival (PFS; 4.1 months vs. 1.8 months, p = .0058). Eight of 10 patients significantly upregulated programmed cell death ligand 1 (PD-L1) on CTCs/CAMLs with treatment (p = .0012). These observations support the safety of the Bria-IMT regimen, demonstrate clinical regressions, imply a role for HLA matching, and identify a possible value for monitoring CAMLs in peripheral blood.
Collapse
Affiliation(s)
| | - Jarrod P. Holmes
- Hematology Oncology, Providence Medical Group Santa Rosa - Cancer Center, Santa Rosa, CA, USA
| | - Carmen Calfa
- Medical Oncology, University of Miami, Miami, FL, USA
| | | | | | | | | | | | - Alex Kharazi
- Development, BriaCell Therapeutics Corp, Philadelphia, PA, USA
- Discovery, Stemedica Cell Technologies, Inc, San Diego, USA
| | - Giuseppe Del Priore
- Development, BriaCell Therapeutics Corp, Philadelphia, PA, USA
- Obstetrics & Gynecology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Mingjin Chang
- Development, BriaCell Therapeutics Corp, Philadelphia, PA, USA
| | | | | |
Collapse
|
2
|
Zhang SW, Wang H, Ding XH, Xiao YL, Shao ZM, You C, Gu YJ, Jiang YZ. Bidirectional crosstalk between therapeutic cancer vaccines and the tumor microenvironment: Beyond tumor antigens. FUNDAMENTAL RESEARCH 2023; 3:1005-1024. [PMID: 38933006 PMCID: PMC11197801 DOI: 10.1016/j.fmre.2022.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 11/20/2022] Open
Abstract
Immunotherapy has rejuvenated cancer therapy, especially after anti-PD-(L)1 came onto the scene. Among the many therapeutic options, therapeutic cancer vaccines are one of the most essential players. Although great progress has been made in research on tumor antigen vaccines, few phase III trials have shown clinical benefits. One of the reasons lies in obstruction from the tumor microenvironment (TME). Meanwhile, the therapeutic cancer vaccine reshapes the TME in an ambivalent way, leading to immune stimulation or immune escape. In this review, we summarize recent progress on the interaction between therapeutic cancer vaccines and the TME. With respect to vaccine resistance, innate immunosuppressive TME components and acquired resistance caused by vaccination are both involved. Understanding the underlying mechanism of this crosstalk provides insight into the treatment of cancer by directly targeting the TME or synergizing with other therapeutics.
Collapse
Affiliation(s)
- Si-Wei Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Han Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiao-Hong Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
3
|
Ayegbusi OS, Enye LA, Saka OS, Omoaghe AO. Counteractive effects of extracts of Mangifera indica on testes of Wistar Rat exposed to cyclophosphamide. Heliyon 2023; 9:e19445. [PMID: 37674830 PMCID: PMC10477475 DOI: 10.1016/j.heliyon.2023.e19445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction Infertility may have a variety of causes that can affect both the male and female reproductive systems. Cyclophosphamide is a drug used in chemotherapy and immune system suppression. Leaf extracts of Mangifera indica exhibit a wide spectrum of pharmacological properties which have been shown in studies, including antioxidant and protective advantages. This study evaluate the antagonistic implications of leaf extracts of Mangifera indica on the testis following the exposure to cyclophosphamide. Methods 25 male Wistar rats were assigned to five groups with five rats in each. Group A (Control), Group B (administered 150 mg of cyclophosphamide only), Group C (administered 50 mg of extracts of leaf extracts of Mangifera indica only), Group D (administered 150 mg of cyclophosphamide and 50 mg of leaf extracts of Mangifera indica) and Group E (administered 150 mg of cyclophosphamide and 100 mg of leaf extracts of Mangifera indica) for two weeks. The rats were euthanized under the anesthetic of ketamine (30 mg/kg IP). Blood was taken by cardiac puncture for biochemical examination. Testes were excised, preserved in 10% Neutral Buffered Formalin for histological investigation. One-way analysis of variance was used to examine the data, and then the Student Newman-Keul post-hoc analysis was performed. The significance of the result was assessed using p < 0.05. Results The study showed statistically significant differences (p < 0.05) in the hormonal assay, including LH, FSH, and testosterone across all test groups, with group B (cyclophosphamide only) having significantly lower levels. Cyclophosphamide administration was observed to have a negative effect on the testicular histology and immunohistochemical results and leaf extracts of Mangifera indica attenuated the damage induced by cyclophosphamide in groups D and E. Conclusion Leaf Extracts of Mangifera indica considerably reduced the effects of cyclophosphamide-induced changes in testis.
Collapse
Affiliation(s)
- Olateju Seun Ayegbusi
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado Ekiti, Nigeria
| | - Linus Anderson Enye
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado Ekiti, Nigeria
| | - Olusola Stephen Saka
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado Ekiti, Nigeria
| | - Adams O. Omoaghe
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado Ekiti, Nigeria
| |
Collapse
|
4
|
Chehelgerdi M, Chehelgerdi M. The use of RNA-based treatments in the field of cancer immunotherapy. Mol Cancer 2023; 22:106. [PMID: 37420174 PMCID: PMC10401791 DOI: 10.1186/s12943-023-01807-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 07/09/2023] Open
Abstract
Over the past several decades, mRNA vaccines have evolved from a theoretical concept to a clinical reality. These vaccines offer several advantages over traditional vaccine techniques, including their high potency, rapid development, low-cost manufacturing, and safe administration. However, until recently, concerns over the instability and inefficient distribution of mRNA in vivo have limited their utility. Fortunately, recent technological advancements have mostly resolved these concerns, resulting in the development of numerous mRNA vaccination platforms for infectious diseases and various types of cancer. These platforms have shown promising outcomes in both animal models and humans. This study highlights the potential of mRNA vaccines as a promising alternative approach to conventional vaccine techniques and cancer treatment. This review article aims to provide a thorough and detailed examination of mRNA vaccines, including their mechanisms of action and potential applications in cancer immunotherapy. Additionally, the article will analyze the current state of mRNA vaccine technology and highlight future directions for the development and implementation of this promising vaccine platform as a mainstream therapeutic option. The review will also discuss potential challenges and limitations of mRNA vaccines, such as their stability and in vivo distribution, and suggest ways to overcome these issues. By providing a comprehensive overview and critical analysis of mRNA vaccines, this review aims to contribute to the advancement of this innovative approach to cancer treatment.
Collapse
Affiliation(s)
- Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
5
|
Zhang Y, Zhu T, Xu S, Gu P, Cai G, Peng S, Liu Z, Yang Y, Hu Y, Liu J, Wang D. Cationic Nanoparticle-Stabilized Vaccine Delivery System for the H9N2 Vaccine to Promote Immune Response in Chickens. Mol Pharm 2023; 20:1613-1623. [PMID: 36795759 DOI: 10.1021/acs.molpharmaceut.2c00805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Chinese yam polysaccharides (CYPs) have received wide attention for their immunomodulatory activity. Our previous studies had discovered that the Chinese yam polysaccharide PLGA-stabilized Pickering emulsion (CYP-PPAS) can serve as an efficient adjuvant to trigger powerful humoral and cellular immunity. Recently, positively charged nano-adjuvants are easily taken up by antigen-presenting cells, potentially resulting in lysosomal escape, the promotion of antigen cross-presentation, and the induction of CD8 T-cell response. However, reports on the practical application of cationic Pickering emulsions as adjuvants are very limited. Considering the economic damage and public-health risks caused by the H9N2 influenza virus, it is urgent to develop an effective adjuvant for boosting humoral and cellular immunity against influenza virus infection. Here, we applied polyethyleneimine-modified Chinese yam polysaccharide PLGA nanoparticles as particle stabilizers and squalene as the oil core to fabricate a positively charged nanoparticle-stabilized Pickering emulsion adjuvant system (PEI-CYP-PPAS). The cationic Pickering emulsion of PEI-CYP-PPAS was utilized as an adjuvant for the H9N2 Avian influenza vaccine, and the adjuvant activity was compared with the Pickering emulsion of CYP-PPAS and the commercial adjuvant (aluminum adjuvant). The PEI-CYP-PPAS, with a size of about 1164.66 nm and a ζ potential of 33.23 mV, could increase the H9N2 antigen loading efficiency by 83.99%. After vaccination with Pickering emulsions based on H9N2 vaccines, PEI-CYP-PPAS generated higher HI titers and stronger IgG antibodies than CYP-PPAS and Alum and increased the immune organ index of the spleen and bursa of Fabricius without immune organ injury. Moreover, treatment with PEI-CYP-PPAS/H9N2 induced CD4+ and CD8+ T-cell activation, a high lymphocyte proliferation index, and increased cytokine expression of IL-4, IL-6, and IFN-γ. Thus, compared with the CYP-PPAS and aluminum adjuvant, the cationic nanoparticle-stabilized vaccine delivery system of PEI-CYP-PPAS was an effective adjuvant for H9N2 vaccination to elicit powerful humoral and cellular immune responses.
Collapse
Affiliation(s)
- Yue Zhang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Tianyu Zhu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Shuwen Xu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Pengfei Gu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Gaofeng Cai
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Song Peng
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Zhenguang Liu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yang Yang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yuanliang Hu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Jiaguo Liu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Deyun Wang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| |
Collapse
|
6
|
Borgovan T, Yanamandra N, Schmidt H. INNATE IMMUNITY AS A TARGET FOR NOVEL THERAPEUTICS IN TRIPLE NEGATIVE BREAST CANCER. Expert Opin Investig Drugs 2022; 31:781-794. [DOI: 10.1080/13543784.2022.2096005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Theo Borgovan
- Oncology Research and DevelopmentGlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426,
| | - Niranjan Yanamandra
- Immuno-Oncology & Combinations Research Unit.GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426,
| | - Hank Schmidt
- Oncology Research and DevelopmentGlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426,
| |
Collapse
|
7
|
Corti C, Giachetti PPMB, Eggermont AMM, Delaloge S, Curigliano G. Therapeutic vaccines for breast cancer: Has the time finally come? Eur J Cancer 2022; 160:150-174. [PMID: 34823982 PMCID: PMC8608270 DOI: 10.1016/j.ejca.2021.10.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
The ability to exploit the immune system as a weapon against cancer has revolutionised the treatment of cancer patients, especially through immune checkpoint inhibitors (ICIs). However, ICIs demonstrated a modest benefit in treating breast cancer (BC), with the exception of certain subsets of triple-negative BCs. An immune-suppressive tumour microenvironment (TME), typically present in BC, is an important factor in the poor response to immunotherapy. After almost two decades of poor clinical trial results, cancer vaccines (CVs), an active immunotherapy, have come back in the spotlight because of some technological advancements, ultimately boosted by coronavirus disease 2019 pandemic. In particular, neoantigens are emerging as the preferred targets for CVs, with gene-based and viral vector-based platforms in development. Moreover, lipid nanoparticles proved to be immunogenic and efficient delivery vehicles. Past clinical trials investigating CVs focused especially on the metastatic disease, where the TME is more likely compromised by inhibitory mechanisms. In this sense, favouring the use of CVs as monotherapy in premalignant or in the adjuvant setting and establishing combination treatments (i.e. CV plus ICI) in late-stage disease are promising strategies. This review provides a full overview of the past and current breast cancer vaccine landscape.
Collapse
Affiliation(s)
- Chiara Corti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Pier P M B Giachetti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Alexander M M Eggermont
- Princess Máxima Center, Utrecht, the Netherlands; Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Suzette Delaloge
- Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
8
|
Wiseman CL, Kharazi A, Sunkari VG, Galeas JL, Dozio V, Hashwah H, Macúchová E, Williams WV, Lacher MD. Regression of Breast Cancer Metastases Following Treatment with Irradiated SV-BR-1-GM, a GM-CSF Overexpressing Breast Cancer Cell Line: Intellectual Property and Immune Markers of Response. Recent Pat Anticancer Drug Discov 2022; 18:224-240. [PMID: 35593340 PMCID: PMC10009895 DOI: 10.2174/1574892817666220518123331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/28/2022] [Accepted: 03/06/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND SV-BR-1-GM, derived from a patient with grade 2 (moderately differentiated) breast cancer, is a GM-CSF-secreting breast cancer cell line with properties of antigen-presenting cells. SV-BR-1-GM and next-generation versions are covered by several pending and granted patents. METHODS We report findings from an open-label phase I, single-arm pilot study with irradiated SV-BR-1-GM cells in 3 breast and 1 ovarian cancer subjects. Inoculations were preceded by lowdose intravenous cyclophosphamide and followed by interferon-alpha2b injections into the SVBR- 1-GM inoculation sites. We assessed both cellular and humoral immune responses, and measured expression levels of SV-BR-1-GM HLA alleles. RESULTS Treatment was generally safe and well tolerated. Immune responses were elicited universally. Overall survival was more than 33 months for three of the four patients. As previously reported, one patient had prompt regression of metastases in lung, breast, and soft tissue. Following cessation of treatment, the patient relapsed widely, including in the brain. Upon retreatment, rapid tumor response was again seen, including complete regression of brain metastases. Consistent with a role of Class II HLA in contributing to SV-BR-1-GM's mechanism of action, this patient allele-matched SV-BR-1-GM at the HLA-DRB1 and HLA-DRB3 loci. We are in the process of developing next-generation SV-BR-1-GM, expressing patient-specific HLAs. Patent applications were filed in various jurisdictions. Thus far, one is granted, in Japan. CONCLUSION A whole-cell immunotherapy regimen with SV-BR-1-GM cells induced regression of metastatic breast cancer. We develop intellectual property based on SV-BR-1-GM's predicted mechanism of action to develop additional whole-cell immunotherapies for cancer patients.
Collapse
Affiliation(s)
- Charles L. Wiseman
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Alexander Kharazi
- Immunotherapy Laboratory, St. Vincent Medical Center, Los Angeles, CA, USA
| | - Vivekananda G. Sunkari
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Jacqueline L. Galeas
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Vito Dozio
- Operations Department, Biognosys AG, Wagistrasse 21, 8952, Schlieren, Switzerland
| | - Hind Hashwah
- Sales and Marketing Nebion AG, Hohlstrasse 515, 8048, Zurich, Switzerland
| | - Eva Macúchová
- Sales and Marketing Nebion AG, Hohlstrasse 515, 8048, Zurich, Switzerland
| | - William V. Williams
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Markus D. Lacher
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| |
Collapse
|
9
|
Williams MM, Hafeez SA, Christenson JL, O’Neill KI, Hammond NG, Richer JK. Reversing an Oncogenic Epithelial-to-Mesenchymal Transition Program in Breast Cancer Reveals Actionable Immune Suppressive Pathways. Pharmaceuticals (Basel) 2021; 14:ph14111122. [PMID: 34832904 PMCID: PMC8622696 DOI: 10.3390/ph14111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Approval of checkpoint inhibitors for treatment of metastatic triple negative breast cancer (mTNBC) has opened the door for the use of immunotherapies against this disease. However, not all patients with mTNBC respond to current immunotherapy approaches such as checkpoint inhibitors. Recent evidence demonstrates that TNBC metastases are more immune suppressed than primary tumors, suggesting that combination or additional immunotherapy strategies may be required to activate an anti-tumor immune attack at metastatic sites. To identify other immune suppressive mechanisms utilized by mTNBC, our group and others manipulated oncogenic epithelial-to-mesenchymal transition (EMT) programs in TNBC models to reveal differences between this breast cancer subtype and its more epithelial counterpart. This review will discuss how EMT modulation revealed several mechanisms, including tumor cell metabolism, cytokine milieu and secretion of additional immune modulators, by which mTNBC cells may suppress both the innate and adaptive anti-tumor immune responses. Many of these pathways/proteins are under preclinical or clinical investigation as therapeutic targets in mTNBC and other advanced cancers to enhance their response to chemotherapy and/or checkpoint inhibitors.
Collapse
|
10
|
Liu Y, Teng L, Fu S, Wang G, Li Z, Ding C, Wang H, Bi L. Highly heterogeneous-related genes of triple-negative breast cancer: potential diagnostic and prognostic biomarkers. BMC Cancer 2021; 21:644. [PMID: 34053447 PMCID: PMC8165798 DOI: 10.1186/s12885-021-08318-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a highly heterogeneous subtype of breast cancer, showing aggressive clinical behaviors and poor outcomes. It urgently needs new therapeutic strategies to improve the prognosis of TNBC. Bioinformatics analyses have been widely used to identify potential biomarkers for facilitating TNBC diagnosis and management. Methods We identified potential biomarkers and analyzed their diagnostic and prognostic values using bioinformatics approaches. Including differential expression gene (DEG) analysis, Receiver Operating Characteristic (ROC) curve analysis, functional enrichment analysis, Protein-Protein Interaction (PPI) network construction, survival analysis, multivariate Cox regression analysis, and Non-negative Matrix Factorization (NMF). Results A total of 105 DEGs were identified between TNBC and other breast cancer subtypes, which were regarded as heterogeneous-related genes. Subsequently, the KEGG enrichment analysis showed that these genes were significantly enriched in ‘cell cycle’ and ‘oocyte meiosis’ related pathways. Four (FAM83B, KITLG, CFD and RBM24) of 105 genes were identified as prognostic signatures in the disease-free interval (DFI) of TNBC patients, as for progression-free interval (PFI), five genes (FAM83B, EXO1, S100B, TYMS and CFD) were obtained. Time-dependent ROC analysis indicated that the multivariate Cox regression models, which were constructed based on these genes, had great predictive performances. Finally, the survival analysis of TNBC subtypes (mesenchymal stem-like [MSL] and mesenchymal [MES]) suggested that FAM83B significantly affected the prognosis of patients. Conclusions The multivariate Cox regression models constructed from four heterogeneous-related genes (FAM83B, KITLG, RBM24 and S100B) showed great prediction performance for TNBC patients’ prognostic. Moreover, FAM83B was an important prognostic feature in several TNBC subtypes (MSL and MES). Our findings provided new biomarkers to facilitate the targeted therapies of TNBC and TNBC subtypes. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08318-1.
Collapse
Affiliation(s)
- Yiduo Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Linxin Teng
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Shiyi Fu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Guiyang Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Zhengjun Li
- College of Health Economics Management, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Chao Ding
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Haodi Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China
| | - Lei Bi
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
11
|
Gordon B, Gadi VK. The Role of the Tumor Microenvironment in Developing Successful Therapeutic and Secondary Prophylactic Breast Cancer Vaccines. Vaccines (Basel) 2020; 8:vaccines8030529. [PMID: 32937885 PMCID: PMC7565925 DOI: 10.3390/vaccines8030529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer affects roughly one in eight women over their lifetime and is a leading cause of cancer-related death in women. While outcomes have improved in recent years, prognosis remains poor for patients who present with either disseminated disease or aggressive molecular subtypes. Cancer immunotherapy has revolutionized the treatment of several cancers, with therapeutic vaccines aiming to direct the cytotoxic immune program against tumor cells showing particular promise. However, these results have yet to translate to breast cancer, which remains largely refractory from such approaches. Recent evidence suggests that the breast tumor microenvironment (TME) is an important and long understudied barrier to the efficacy of therapeutic vaccines. Through an improved understanding of the complex and biologically diverse breast TME, it may be possible to advance new combination strategies to render breast carcinomas sensitive to the effects of therapeutic vaccines. Here, we discuss past and present efforts to advance therapeutic vaccines in the treatment of breast cancer, the molecular mechanisms through which the TME contributes to the failure of such approaches, as well as the potential means through which these can be overcome.
Collapse
Affiliation(s)
- Benjamin Gordon
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Correspondence:
| | - Vijayakrishna K. Gadi
- Division of Hematology and Oncology, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
12
|
Guo X, Zhang T, Wang X, Su H, Sun W, Liu Y, Kang K, Liu T, Jiang S, Wang Y, Wang D, Yin H, Tian L, Li D, Ren G. The immune enhancement effects of recombinant NDV expressing chicken granulocyte-macrophage colony-stimulating factor on the different avian influenza vaccine subtypes. Transbound Emerg Dis 2020; 67:2108-2117. [PMID: 32246561 DOI: 10.1111/tbed.13559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/05/2020] [Accepted: 03/14/2020] [Indexed: 01/02/2023]
Abstract
Avian influenza is an acute and highly contagious infectious disease that is caused by the influenza virus. Avian influenza has been widely spread all over the world, has caused property loss and has threatened human life and security. In this study, the recombinant plasmid rClone30-chGM-CSF was constructed and rescued to the recombinant virus rClone30-chGM-CSF successfully. After 8 days of immunization with the recombinant virus, the titre of NDV HI (haemagglutination inhibition) antibodies in SPF chickens reached its peak. The average titre of the rClone30-chGM-CSF group reached 6 log2 and was significantly higher than the protection critical value of 4 log2 ; the titres of the rClone30 group and the blank group were 2.86 log2 and 1 log2 , respectively, indicating that the recombinant virus can effectively improve the NDV antibody titre. Then, SPF chickens were co-immunized with the recombinant virus and with three different vaccine subtypes of inactivated avian influenza. The results indicated that the SPF chickens that were immunized with the vaccine plus rClone30-chGM-CSF showed significantly higher avian influenza antibody levels than those in the single vaccine groups. Furthermore, the SPF chickens in the vaccine plus rClone30-chGM-CSF group elicited stronger CD4+ and CD8+ T-cell proliferative responses and also had upregulated transcriptional levels of interleukin-1β (IL-1β), IL-4, IL-6 and IL-17 compared with those in the single vaccine groups. This study has shown that the recombinant virus expressing chicken granulocyte-macrophage colony-stimulating factor (chGM-CSF) can be used not only as an NDV vaccine to effectively improve the titre of NDV antibodies but also as a biological adjuvant to enhance the immune effects of the avian influenza vaccine. Therefore, this recombinant virus can also be used as a biological adjuvant for other poultry vaccines.
Collapse
Affiliation(s)
- Xiaochen Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Teng Zhang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xiangxiang Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Han Su
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Wenying Sun
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yunye Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Kai Kang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Tianyan Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Shan Jiang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yaoqun Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Dan Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - He Yin
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Limin Tian
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Deshan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
- Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin, China
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
- Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin, China
| |
Collapse
|
13
|
Quantification and characterization of granulocyte macrophage colony-stimulating factor activated human peripheral blood mononuclear cells by fluorine-19 cellular MRI in an immunocompromised mouse model. Diagn Interv Imaging 2020; 101:577-588. [DOI: 10.1016/j.diii.2020.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
|
14
|
Okoli UA, Okafor MT, Agu KA, Ndubuisi AC, Nwigwe IJ, Nna EO, Okafor OC, Ukekwe FI, Nwagha TU, Menkiti VC, Eze CO, Onyekwelu KC, Ikekpeazu JE, Anusiem CA, Mbah AU, Chijioke CP, Udeniya IJ. Methodology for processing mastectomy and cryopreservation of breast cancer tissue in a resource- poor setting: A pilot study. Cryobiology 2020; 97:179-184. [PMID: 32562613 DOI: 10.1016/j.cryobiol.2020.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND There is scarcity of breast cancer tissues derived from women of African origin available for patient - derived xenograft and organoid models. OBJECTIVE We aim to create a versatile protocol for processing mastectomy and cryopreservation of breast cancer tissue. METHODOLOGY An immediate collection of breast cancer tissue from mastectomy was bathed in 4 °C HBSS and immediately transferred to 4 °C RPMI1640 containing HEPES, 10% FBS, Streptomycin and Penicillin. Tissues were processed over ice yielding nine samples of cold ischemic time (20-45 min) stored at 3 min interval. Cut samples were transferred into cryovials containing 4 °C cryoprotectant agent (90% FBS +10% Me2SO) before snap -freezing in liquid Nitrogen vapour and final short-term storage in -80 °C Freezer. The histomorphology, tissue and molecular viability were assessed. RESULTS The cold ischemic times had no detrimental effect to the nine samples despite being processed in a resource poor setting, hence providing a reproducible and reliable protocol.
Collapse
Affiliation(s)
- Uzoamaka A Okoli
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria; Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria Enugu Campus, Nigeria.
| | - Michael T Okafor
- Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria Enugu Campus, Nigeria.
| | - Kenneth A Agu
- Department of Surgery, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Augustine C Ndubuisi
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Ifeoma J Nwigwe
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Emmanuel O Nna
- Molecular Pathology Institute, Rangers Avenue, Independence Layout, Enugu, Nigeria
| | - Okechukwu C Okafor
- Department of Morbid Anatomy, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Francis I Ukekwe
- Department of Morbid Anatomy, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Teresa U Nwagha
- Department of Haematology and Immunology, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Victor C Menkiti
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria; Cleon Healthcare Laboratory, G.R.A, Enugu, Nigeria
| | - Charles O Eze
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Kenechukwu C Onyekwelu
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Joy E Ikekpeazu
- Department of Medical Biochemistry and Molecular Biology, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Chikere A Anusiem
- Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Anthony U Mbah
- Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Chioli P Chijioke
- Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| | - Iroka J Udeniya
- Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria Enugu Campus, Nigeria
| |
Collapse
|
15
|
Xu Y, Zou R, Wang J, Wang ZW, Zhu X. The role of the cancer testis antigen PRAME in tumorigenesis and immunotherapy in human cancer. Cell Prolif 2020; 53:e12770. [PMID: 32022332 PMCID: PMC7106952 DOI: 10.1111/cpr.12770] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/01/2020] [Accepted: 01/15/2020] [Indexed: 12/24/2022] Open
Abstract
Preferentially expressed antigen in melanoma (PRAME), which belongs to the cancer/testis antigen (CTA) gene family, plays a pivotal role in multiple cellular processes and immunotherapy response in human cancers. PRAME is highly expressed in different types of cancers and is involved in cell proliferation, apoptosis, differentiation and metastasis as well as the outcomes of patients with cancer. In this review article, we discuss the potential roles and physiological functions of PRAME in various types of cancers. Moreover, this review highlights immunotherapeutic strategies that target PRAME in human malignancies. Therefore, the modulation of PRAME might be useful for the treatment of patients with cancer.
Collapse
Affiliation(s)
- Yichi Xu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruanmin Zou
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Wei Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Chun BM, Page DB, McArthur HL. Combination Immunotherapy Strategies in Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00333-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
We summarize combination immunotherapy strategies for the treatment of breast cancer, with a focus on metastatic disease. First, a general overview of combination approaches is presented according to breast cancer subtype. Second, additional review of promising combination approaches is presented.
Recent Findings
Combination strategies utilizing chemotherapy or radiotherapy with immune checkpoint inhibition are being evaluated across multiple phase III trials. Dual immunotherapy strategies, such as dual immune checkpoint inhibition or combined co-stimulation/co-inhibition, have supportive preclinical evidence and are under early clinical investigation. Modulation of the immune microenvironment via cytokines and vaccination strategies, as well as locally focused treatments to enhance antigenic responses, are active areas of research.
Summary
Pre-clinical and translational research sheds new light on numerous ways the immune system may be modulated to fight against cancer. We describe current and emerging combination approaches which may improve patient outcomes in metastatic breast cancer.
Collapse
|
17
|
Mengos AE, Gastineau DA, Gustafson MP. The CD14 +HLA-DR lo/neg Monocyte: An Immunosuppressive Phenotype That Restrains Responses to Cancer Immunotherapy. Front Immunol 2019; 10:1147. [PMID: 31191529 PMCID: PMC6540944 DOI: 10.3389/fimmu.2019.01147] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022] Open
Abstract
Recent successes in cancer immunotherapy have been tempered by sub-optimal clinical responses in the majority of patients. The impaired anti-tumor immune responses observed in these patients are likely a consequence of immune system dysfunction contributed to by a variety of factors that include, but are not limited to, diminished antigen presentation/detection, leukopenia, a coordinated network of immunosuppressive cell surface proteins, cytokines and cellular mediators. Monocytes that have diminished or no HLA-DR expression, called CD14+HLA-DRlo/neg monocytes, have emerged as important mediators of tumor-induced immunosuppression. These cells have been grouped into a larger class of suppressive cells called myeloid derived suppressor cells (MDSCs) and are commonly referred to as monocytic myeloid derived suppressor cells. CD14+HLA-DRlo/neg monocytes were first characterized in patients with sepsis and were shown to regulate the transition from the inflammatory state to immune suppression, ultimately leading to immune paralysis. These immunosuppressive monocytes have also recently been shown to negatively affect responses to PD-1 and CTLA-4 checkpoint inhibition, CAR-T cell therapy, cancer vaccines, and hematopoietic stem cell transplantation. Ultimately, the goal is to understand the role of these cells in the context of immunosuppression not only to facilitate the development of targeted therapies to circumvent their effects, but also to potentially use them as a biomarker for understanding disparate responses to immunotherapeutic regimens. Practical aspects to be explored for development of CD14+HLA-DRlo/neg monocyte detection in patients are the standardization of flow cytometric gating methods to assess HLA-DR expression, an appropriate quantitation method, test sample type, and processing guidances. Once detection methods are established that yield consistently reproducible results, then further progress can be made toward understanding the role of CD14+HLA-DRlo/neg monocytes in the immunosuppressive state.
Collapse
Affiliation(s)
- April E Mengos
- Nyberg Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, United States
| | - Dennis A Gastineau
- Nyberg Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, United States
| | - Michael P Gustafson
- Nyberg Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|