1
|
Goodarzi MM, Mosayebi G, Ganji A, Raoufi E, Sadelaji S, Babaei S, Abtahi H. HPV16 mutant E6/E7 construct is protective in mouse model. BMC Biotechnol 2024; 24:71. [PMID: 39350162 PMCID: PMC11443707 DOI: 10.1186/s12896-024-00893-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Human papillomavirus type 16 (HPV-16) infection is strongly associated with considerable parts of cervical, neck, and head cancers. Performed investigations have had moderate clinical success, so research to reach an efficient vaccine has been of great interest. In the present study, the immunization potential of a newly designed HPV-16 construct was evaluated in a mouse model. RESULTS Initially, a construct containing HPV-16 mutant (m) E6/E7 fusion gene was designed and antigen produced in two platforms (i.e., DNA vaccine and recombinant protein). Subsequently, the immunogenicity of these platforms was investigated in five mice) C57BL/6 (groups based on several administration strategies. Three mice groups were immunized recombinant protein, DNA vaccine, and a combination of them, and two other groups were negative controls. The peripheral blood mononuclear cells (PBMCs) proliferation, Interleukin-5 (IL-5) and interferon-γ (IFN-γ) cytokines, IgG1 and IgG2a antibody levels were measured. After two weeks, TC-1 tumor cells were injected into all mice groups, and subsequently further analysis of tumor growth and metastasis and mice survival were performed according to the schedule. Overall, the results obtained from in vitro immunology and tumor cells challenging assays indicated the potential of the mE6/E7 construct as an HPV16 therapeutic vaccine candidate. The results demonstrated a significant increase in IFN-γ cytokine (P value < 0.05) in the Protein/Protein (D) and DNA/Protein (E) groups. This finding was in agreement with in vivo assays. Control groups show a 10.5-fold increase (P value < 0.001) and (C) DNA/DNA group shows a 2.5-fold increase (P value < 0.01) in tumor growth compared to D and E groups. Also, a significant increase in survival of D and E (P value < 0.001) and C (P value < 0.01) groups were observed. CONCLUSIONS So, according to the findings, the recombinant protein could induce stronger protection compared to the DNA vaccine form. Protein/Protein and DNA/Protein are promising administration strategies for presenting this construct to develop an HPV-16 therapeutic vaccine candidate.
Collapse
Affiliation(s)
- Maryam Moazami Goodarzi
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ghasem Mosayebi
- Department of Microbiology and Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ali Ganji
- Department of Microbiology and Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ehsan Raoufi
- Vaccine research center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Sadelaji
- Department of Microbiology and Immunology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Saeid Babaei
- Department of Anatomy, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Hamid Abtahi
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran.
| |
Collapse
|
2
|
Takahashi R, Osumi H, Wakatsuki T, Yamamoto N, Taguchi S, Nakayama I, Ooki A, Ogura M, Takahari D, Chin K, Yamaguchi K, Shinozaki E. Clinical outcomes and prognostic factors of concurrent chemoradiotherapy for anal squamous cell carcinoma in Japan. Int J Clin Oncol 2024; 29:1161-1172. [PMID: 38819609 DOI: 10.1007/s10147-024-02540-0] [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: 01/06/2024] [Accepted: 04/20/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Concurrent chemoradiotherapy (CCRT) is the standard treatment for locoregional anal squamous cell carcinoma (ASCC) in western countries. However, there have been few reports on the clinical outcomes of CCRT in Japan. This study aimed to evaluate the clinical outcomes of CCRT, prognostic factors, and the clinical impact of programmed cell death-ligand 1 (PD-L1) expression of ASCC in Japan. METHODS Patients with locoregional ASCC were enrolled between 2007 and 2017. All patients received CCRT consisting of ≥ 45 Gy of radiation, 5-fluorouracil, and mitomycin C. Disease-free survival (DFS), overall survival (OS), and adverse events (AEs) were estimated. Expression of p16 and PD-L1 were assessed by immunohistochemical staining (IHC). RESULTS This study included 36 patients, of whom 30 (83.3%) were female. Among the participants, 32 (88.9%) achieved complete clinical remission, while six (16.7%) experienced recurrence. The five-year DFS and five-year OS were 72.2% and 84.7%, respectively. Grades ≥ 3 serious AEs included neutropenia in 10 (27.7%) and perianal dermatitis in eight (22.2%). In a univariate analysis, male sex, lymph node metastasis, and large tumor size were significantly associated with worse outcome. In a multivariate analysis, tumor size was an independent factor associated with short DFS. Of the 30 patients whose biopsy specimens were available for IHC, 29 (96.7%) were positive for p16, and 13 (43.3%) were positive for PD-L1. However, PD-L1 expression did not show any clinical impact. CONCLUSIONS The comparative etiology, clinical outcomes, and prognostic factors of CCRT observed in Japanese patients with locoregional ASCC were consistent with western data.
Collapse
Affiliation(s)
- Ryo Takahashi
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of General Surgery, Heiman Municipal Hospital, Aichi, Japan
| | - Hiroki Osumi
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takeru Wakatsuki
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Noriko Yamamoto
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Senzo Taguchi
- Department of Radiation Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Izuma Nakayama
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akira Ooki
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Mariko Ogura
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Daisuke Takahari
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Keisho Chin
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Eiji Shinozaki
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| |
Collapse
|
3
|
Lo Cigno I, Calati F, Girone C, Catozzo M, Gariglio M. High-risk HPV oncoproteins E6 and E7 and their interplay with the innate immune response: Uncovering mechanisms of immune evasion and therapeutic prospects. J Med Virol 2024; 96:e29685. [PMID: 38783790 DOI: 10.1002/jmv.29685] [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: 02/20/2024] [Revised: 04/22/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
Human papillomaviruses (HPVs) are double-stranded DNA (dsDNA) tumor viruses causally associated with 5% of human cancers, comprising both anogenital and upper aerodigestive tract carcinomas. Despite the availability of prophylactic vaccines, HPVs continue to pose a significant global health challenge, primarily due to inadequate vaccine access and coverage. These viruses can establish persistent infections by evading both the intrinsic defenses of infected tissues and the extrinsic defenses provided by professional innate immune cells. Crucial for their evasion strategies is their unique intraepithelial life cycle, which effectively shields them from host detection. Thus, strategies aimed at reactivating the innate immune response within infected or transformed epithelial cells, particularly through the production of type I interferons (IFNs) and lymphocyte-recruiting chemokines, are considered viable solutions to counteract the adverse effects of persistent infections by these oncogenic viruses. This review focuses on the complex interplay between the high-risk HPV oncoproteins E6 and E7 and the innate immune response in epithelial cells and HPV-associated cancers. In particular, it details the molecular mechanisms by which E6 and E7 modulate the innate immune response, highlighting significant progress in our comprehension of these processes. It also examines forward-looking strategies that exploit the innate immune system to ameliorate existing anticancer therapies, thereby providing crucial insights into future therapeutic developments.
Collapse
Affiliation(s)
- Irene Lo Cigno
- Virology Unit, Department of Translational Medicine, Eastern Piedmont University, Novara, Italy
| | - Federica Calati
- Virology Unit, Department of Translational Medicine, Eastern Piedmont University, Novara, Italy
| | - Carlo Girone
- Virology Unit, Department of Translational Medicine, Eastern Piedmont University, Novara, Italy
| | - Marta Catozzo
- Virology Unit, Department of Translational Medicine, Eastern Piedmont University, Novara, Italy
| | - Marisa Gariglio
- Virology Unit, Department of Translational Medicine, Eastern Piedmont University, Novara, Italy
| |
Collapse
|
4
|
Trammel J, Amusan O, Hultgren A, Raikhy G, Bodily JM. Epidermal growth factor receptor-dependent stimulation of differentiation by human papillomavirus type 16 E5. Virology 2024; 590:109952. [PMID: 38103269 PMCID: PMC10842332 DOI: 10.1016/j.virol.2023.109952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/19/2023]
Abstract
Human papillomaviruses (HPVs) infect keratinocytes of stratified squamous epithelia, and persistent infection with high-risk HPV types, such as HPV16, may lead to the development of malignancies. HPV evades host immunity in part by linking its gene expression to the host differentiation program, and therefore relies on differentiation to complete its life cycle. Based on previous reports indicating that the HPV16 protein E5 is important in the late stages of the differentiation-dependent life cycle, we found that organotypic cultures harboring HPV16 genomes lacking E5 showed reduced markers of terminal differentiation compared to wild type HPV16-containing cultures. We found that epidermal growth factor receptor (EGFR) levels and activation were increased in an E5-depdendent manner in these tissues, and that EGFR promoted terminal differentiation and expression of the HPV16 L1 gene. These findings suggest a function for E5 in preserving the ability of HPV16 containing keratinocytes to differentiate, thus facilitating the production of new virus progeny.
Collapse
Affiliation(s)
- Jessica Trammel
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Oluwamuyiwa Amusan
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Allison Hultgren
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA; School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Gaurav Raikhy
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Jason M Bodily
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
5
|
Jesus ACC, Meniconi MCG, Galo LK, Duarte MIS, Sotto MN, Pagliari C. Plasmacytoid Dendritic Cells, the Expression of the Stimulator of Interferon Genes Protein (STING) and a Possible Role of Th17 Immune Response in Cervical Lesions Mediated by Human Papillomavirus. Indian J Microbiol 2023; 63:588-595. [PMID: 38031606 PMCID: PMC10682341 DOI: 10.1007/s12088-023-01117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/07/2023] [Indexed: 12/01/2023] Open
Abstract
Human papillomavirus (HPV) is a virus with a DNA structure that specifically targets squamous epithelial cells. In individuals with a healthy immune system, HPV infection is typically resolved naturally, leading to spontaneous regression. However, when the viral genetic material integrates into the host DNA, it can disrupt the immune response and eventually give rise to neoplastic manifestations. Remarkably, HPV infection appears to activate a protein called Stimulator of Interferon genes (STING), which contributes to the infiltration of Treg Foxp3 + cells. This cellular response acts as a predisposing factor in patients with HPV, potentially exacerbating the progression of the infection. The STING is versatile in different circumstances and can play a role in the immune response as an anti-tumour therapeutic target in HPV-related carcinogenesis. The function of Th17 cells is ambiguous, depending on the microenvironment in the tumour. In this study, 46 biopsies of the uterine cervix of human immunodeficiency virus (HIV) positive patients were divided into three groups: I-cervicitis (10); II-low-grade intraepithelial neoplasia (26); III-moderate or severe intraepithelial neoplasia (10) and it was performed an immunohistochemical technique with the specific antibodies to HPV, CD123, STING and IL17. Immunostained cells were quantified and statistically analysed. Antigens of HPV were detected in the cervical intraepithelial neoplasia (CIN) groups and were absent in cervicitis group. The expression of CD123 was positive in 10.87% of the casuistic, with no statistical difference among groups. STING was present in the three groups. Group 1 presented an area fraction that varied from 3 to 20%, group 2 presented a variation of 3-23% and group 3 presented an area fraction between 4 and 12%. Cells expressing IL17 were present in three groups, more frequent in cervicitis. Considering that the casuistic is composed of women carrying HIV, this infectious agent could influence the numerical similarities of the cells studied among three groups, even in the absence of HPV.
Collapse
Affiliation(s)
- Ana Carolina Caetano Jesus
- Programa de Pós-Graduação em Ciências da Saúde - Instituto de Assistência Médica ao Servidor Público Estadual, São Paulo, SP Brazil
- Departamento de Patologia, Faculdade de Medicina da USP, Universidade de São Paulo, Av Dr Arnaldo, 455 sala 1118, São Paulo, SP CEP 01246-903 Brazil
| | - Maria Cristina Gonçalves Meniconi
- Departamento de Patologia, Faculdade de Medicina da USP, Universidade de São Paulo, Av Dr Arnaldo, 455 sala 1118, São Paulo, SP CEP 01246-903 Brazil
| | - Luciane Kanashiro Galo
- Departamento de Patologia, Faculdade de Medicina da USP, Universidade de São Paulo, Av Dr Arnaldo, 455 sala 1118, São Paulo, SP CEP 01246-903 Brazil
| | - Maria Irma Seixas Duarte
- Departamento de Patologia, Faculdade de Medicina da USP, Universidade de São Paulo, Av Dr Arnaldo, 455 sala 1118, São Paulo, SP CEP 01246-903 Brazil
| | - Mirian Nacagami Sotto
- Departamento de Patologia, Faculdade de Medicina da USP, Universidade de São Paulo, Av Dr Arnaldo, 455 sala 1118, São Paulo, SP CEP 01246-903 Brazil
| | - Carla Pagliari
- Programa de Pós-Graduação em Ciências da Saúde - Instituto de Assistência Médica ao Servidor Público Estadual, São Paulo, SP Brazil
- Departamento de Patologia, Faculdade de Medicina da USP, Universidade de São Paulo, Av Dr Arnaldo, 455 sala 1118, São Paulo, SP CEP 01246-903 Brazil
| |
Collapse
|
6
|
Jiang L, Ma S, Zhang G, Jiang L, Yan L. Analysis of tobacco exposures and high-risk HPV infection in American women: National Health and Nutrition Examination Survey. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:110489-110498. [PMID: 37792188 PMCID: PMC10625505 DOI: 10.1007/s11356-023-30175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023]
Abstract
Nicotine is a known toxin, but its relationship with cervicovaginal high-risk human papillomavirus (HR-HPV) infection is uncertain. This study aimed to investigate whether tobacco exposure is associated with elevated cervicovaginal HR-HPV infection in US women, and if the strength of this association varies with the degree of exposure. Cross-sectional data from the 2011-2016 National Health and Nutrition Examination Survey (NHANES), which included a nationally representative sample of US women, were used for the study. Out of 12436 women aged 18-59 who participated in the interview, 3833 were ultimately enrolled in this study. Weighted logistic regression was used to estimate the link between tobacco exposure and cervicovaginal HR-HPV infection. The mean age of participants was 38.6 (SD 12.1) years, and non-Hispanic White individuals accounted for 37.3% of the sample. Those with any tobacco exposure tended to be younger (mean age 37.7 [SD 12.4] years vs 40.3 [11.2] years), non-Hispanic Black (27.8% vs. 15.1%), lower educated (41.8% vs. 29.4%), and have lower family income (39.9% vs. 23.5%). After adjustment, the odds of having HR-HPV infection were 1.32 (95% CI, 1.09-1.59) for those exposed to tobacco, remaining significant in multiple sensitivity analyses and across subgroups. This study, based on a nationally representative sample from the United States, suggests that tobacco exposure is a risk factor for elevated HR-HPV infection in women, highlighting the need for further research into reducing this modifiable risk factor.
Collapse
Affiliation(s)
- Liangzi Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, Jinan, 250000, China
| | - Suting Ma
- Department of Pediatrics, Linyi People's Hospital, Linyi, 276000, China
| | - Ge Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, Jinan, 250000, China
| | - Lingling Jiang
- Department of Gastroenterology, Shandong Medical College (Linyi Geriatric Hospital), Linyi, 276000, China
| | - Li Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, Jinan, 250000, China.
| |
Collapse
|
7
|
Ahmed H, Mahmud AR, Siddiquee MFR, Shahriar A, Biswas P, Shimul MEK, Ahmed SZ, Ema TI, Rahman N, Khan MA, Mizan MFR, Emran TB. Role of T cells in cancer immunotherapy: Opportunities and challenges. CANCER PATHOGENESIS AND THERAPY 2023; 1:116-126. [PMID: 38328405 PMCID: PMC10846312 DOI: 10.1016/j.cpt.2022.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 09/01/2023]
Abstract
Immunotherapies boosting the immune system's ability to target cancer cells are promising for the treatment of various tumor types, yet clinical responses differ among patients and cancers. Recently, there has been increasing interest in novel cancer immunotherapy practices aimed at triggering T cell-mediated anti-tumor responses. Antigen-directed cytotoxicity mediated by T lymphocytes has become a central focal point in the battle against cancer utilizing the immune system. The molecular and cellular mechanisms involved in the actions of T lymphocytes have directed new therapeutic approaches in cancer immunotherapy, including checkpoint blockade, adoptive and chimeric antigen receptor (CAR) T cell therapy, and cancer vaccinology. This review addresses all the strategies targeting tumor pathogenesis, including metabolic pathways, to evaluate the clinical significance of current and future immunotherapies for patients with cancer, which are further engaged in T cell activation, differentiation, and response against tumors.
Collapse
Affiliation(s)
- Hossain Ahmed
- Department of Biotechnology and Genetic Engineering, University of Development Alternative (UODA), 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | | | - Asif Shahriar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Partha Biswas
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology (JUST), Jashore, 7408, Bangladesh
| | - Md. Ebrahim Khalil Shimul
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology (JUST), Jashore, 7408, Bangladesh
| | - Shahlaa Zernaz Ahmed
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh
| | - Tanzila Ismail Ema
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh
| | - Nova Rahman
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md. Arif Khan
- Department of Biotechnology and Genetic Engineering, University of Development Alternative (UODA), 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | | | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| |
Collapse
|
8
|
Glover A, Zhang Z, Shannon-Lowe C. Deciphering the roles of myeloid derived suppressor cells in viral oncogenesis. Front Immunol 2023; 14:1161848. [PMID: 37033972 PMCID: PMC10076641 DOI: 10.3389/fimmu.2023.1161848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Myeloid derived suppressor cells (MDSCs) are a heterogenous population of myeloid cells derived from monocyte and granulocyte precursors. They are pathologically expanded in conditions of ongoing inflammation where they function to suppress both innate and adaptive immunity. They are subdivided into three distinct subsets: monocytic (M-) MDSC, polymorphonuclear (or neutrophilic) (PMN-) MDSC and early-stage (e-) MDSC that may exhibit differential function in different pathological scenarios. However, in cancer they are associated with inhibition of the anti-tumour immune response and are universally associated with a poor prognosis. Seven human viruses classified as Group I carcinogenic agents are jointly responsible for nearly one fifth of all human cancers. These viruses represent a large diversity of species, including DNA, RNA and retroviridae. They include the human gammaherpesviruses (Epstein Barr virus (EBV) and Kaposi's Sarcoma-Associated Herpesvirus (KSHV), members of the high-risk human papillomaviruses (HPVs), hepatitis B and C (HBV, HCV), Human T cell leukaemia virus (HTLV-1) and Merkel cell polyomavirus (MCPyV). Each of these viruses encode an array of different oncogenes that perturb numerous cellular pathways that ultimately, over time, lead to cancer. A prerequisite for oncogenesis is therefore establishment of chronic infection whereby the virus persists in the host cells without being eradicated by the antiviral immune response. Although some of the viruses can directly modulate the immune response to enable persistence, a growing body of evidence suggests the immune microenvironment is modulated by expansions of MDSCs, driven by viral persistence and oncogenesis. It is likely these MDSCs play a role in loss of immune recognition and function and it is therefore essential to understand their phenotype and function, particularly given the increasing importance of immunotherapy in the modern arsenal of anti-cancer therapies. This review will discuss the role of MDSCs in viral oncogenesis. In particular we will focus upon the mechanisms thought to drive the MDSC expansions, the subsets expanded and their impact upon the immune microenvironment. Importantly we will explore how MDSCs may modulate current immunotherapies and their impact upon the success of future immune-based therapies.
Collapse
|
9
|
Maiorano BA, Maiorano MFP, Ciardiello D, Maglione A, Orditura M, Lorusso D, Maiello E. Beyond Platinum, ICIs in Metastatic Cervical Cancer: A Systematic Review. Cancers (Basel) 2022; 14:cancers14235955. [PMID: 36497437 PMCID: PMC9737392 DOI: 10.3390/cancers14235955] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Cervical cancer (CC) constitutes the fourth most common tumor among the female population. Therapeutic approaches to advanced CC are limited, with dismal results in terms of survival, mainly after progression to platinum-based regimens. Immune checkpoint inhibitors (ICIs) are remodeling the therapeutic scenario of many solid tumors. The role of ICIs in CC should be addressed. Therefore, we systematically reviewed the latest clinical trials employing ICIs in advanced CC to assess which ICIs have been employed and how ICIs might meet the need for new therapeutic options in terms of efficacy and safety. METHODS The review was conducted following the PRISMA guidelines. The following efficacy outcomes were specifically collected: overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS); for safety: type, number, and grade of adverse events (AEs). RESULTS A total of 17 studies were analyzed. Anti-PD1 (pembrolizumab, nivolumab, cemiplimab, balstilimab, and tislelizumab), anti-PD-L1 (atezolizumab), and anti-CTLA-4 (ipilimumab, zalifrelimab) agents were employed both as single agents or combinations. Overall ORR ranged from 0% to 65.9%. ORR ranged from 5.9% to 69.6% in PD-L1-positive patients and from 0% to 50% in PD-L1-negative patients. DCR was 30.6-94.1%. mPFS ranged from 2 to 10.4 months. mOS ranged from 8 months to not reached. PD-L1 status did not impact survival. A total of 33.9% to 100% of patients experienced AEs. CONCLUSION Immunotherapy represents an appealing strategy for patients with advanced CC, as 2 out of 3 patients seem to respond to ICIs. PD-L1 status might be an indicator of response without impacting survival.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Mauro Francesco Pio Maiorano
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence:
| | - Davide Ciardiello
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Annamaria Maglione
- Obstetrics and Gynecology Department, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
| | - Michele Orditura
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Domenica Lorusso
- Department of Women and Child Health, Division of Gynaecologic Oncology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
10
|
Enhancing the Effect of Nucleic Acid Vaccines in the Treatment of HPV-Related Cancers: An Overview of Delivery Systems. Pathogens 2022; 11:pathogens11121444. [PMID: 36558778 PMCID: PMC9781236 DOI: 10.3390/pathogens11121444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Prophylactic vaccines against human papillomavirus (HPV) have proven efficacy in those who have not been infected by the virus. However, they do not benefit patients with established tumors. Therefore, the development of therapeutic options for HPV-related malignancies is critical. Third-generation vaccines based on nucleic acids are fast and simple approaches to eliciting adaptive immune responses. However, techniques to boost immunogenicity, reduce degradation, and facilitate their capture by immune cells are frequently required. One option to overcome this constraint is to employ delivery systems that allow selective antigen absorption and help modulate the immune response. This review aimed to discuss the influence of these different systems on the response generated by nucleic acid vaccines. The results indicate that delivery systems based on lipids, polymers, and microorganisms such as yeasts can be used to ensure the stability and transport of nucleic acid vaccines to their respective protein synthesis compartments. Thus, in view of the limitations of nucleic acid-based vaccines, it is important to consider the type of delivery system to be used-due to its impact on the immune response and desired final effect.
Collapse
|
11
|
Suominen H, Paaso A, Koskimaa HM, Grénman S, Syrjänen K, Syrjänen S, Louvanto K. Peripheral Blood T-lymphocyte Phenotypes in Mother-Child Pairs Stratified by the Maternal HPV Status: Persistent HPV16 vs. HPV-Negative: A Case-Control Study. Viruses 2022; 14:v14122633. [PMID: 36560637 PMCID: PMC9788282 DOI: 10.3390/v14122633] [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: 10/12/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Only few studies exist on the phenotype distribution of peripheral blood lymphocytes concerning persistent oral HPV infection. T-lymphocyte subsets were phenotyped in women who had persistent genital or oral HPV16 infection, using HPV-negative women as a reference group. A subset of 42 mothers and their children (n = 28), were stratified into two groups according to the mothers' HPV status. PBMCs from previously cryopreserved venous samples were immunophenotyped by flow cytometry. Proportions of the CD4+ or CD8+ lymphocytes by their immunophenotype subsets were compared between HPV-positive and -negative mothers and their children. The mean rank distribution of CD8+ memory cells was significantly higher among mothers with persistent genital HPV16 infection. The median levels of both the antigen-presenting CD4+ cells and activated CD8+ cells were significantly lower in mothers with persistent oral HPV16 infection. When oral and genital HPV16-persistors were analyzed as a group, a marker of terminal effector cells was significantly increased as compared to HPV-negative women. Significantly higher levels of activated CD4+, CD8+ and circulating CD8+ memory cells were found among children whose mothers had persistent oral HPV16 infection. Persistent HPV16 infections are associated with changes in peripheral blood T-lymphocyte subsets. The mother's persistent oral HPV16 infection possibly results in immune alterations in her offspring.
Collapse
Affiliation(s)
- Helmi Suominen
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
| | - Anna Paaso
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Hanna-Mari Koskimaa
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Seija Grénman
- Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, 20014 Turku, Finland
| | | | - Stina Syrjänen
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Department of Pathology, Turku University Hospital, 20014 Turku, Finland
| | - Karolina Louvanto
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, 33100 Tampere, Finland
- Correspondence: ; Tel.: +35-8504713838
| |
Collapse
|
12
|
Mo Y, Ma J, Zhang H, Shen J, Chen J, Hong J, Xu Y, Qian C. Prophylactic and Therapeutic HPV Vaccines: Current Scenario and Perspectives. Front Cell Infect Microbiol 2022; 12:909223. [PMID: 35860379 PMCID: PMC9289603 DOI: 10.3389/fcimb.2022.909223] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/06/2022] [Indexed: 12/20/2022] Open
Abstract
Persistent human papillomavirus (HPV) infection is recognized as the main cause of cervical cancer and other malignant cancers. Although early detection and treatment can be achieved by effective HPV screening methods and surgical procedures, the disease load has not been adequately mitigated yet, especially in the underdeveloped areas. Vaccine, being regarded as a more effective solution, is expected to prevent virus infection and the consequent diseases in the phases of both prevention and treatment. Currently, there are three licensed prophylactic vaccines for L1-VLPs, namely bivalent, quadrivalent and nonavalent vaccine. About 90% of HPV infections have been effectively prevented with the implementation of vaccines worldwide. However, no significant therapeutic effect has been observed on the already existed infections and lesions. Therapeutic vaccine designed for oncoprotein E6/E7 activates cellular immunity rather than focuses on neutralizing antibodies, which is considered as an ideal immune method to eliminate infection. In this review, we elaborate on the classification, mechanism, and clinical effects of HPV vaccines for disease prevention and treatment, in order to make improvements to the current situation of HPV vaccines by provoking new ideas.
Collapse
Affiliation(s)
- Yicheng Mo
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiabing Ma
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Hongtao Zhang
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Junjie Shen
- IND Center, Chongqing Precision Biotech Co., Ltd., Chongqing, China
| | - Jun Chen
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Juan Hong
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Yanmin Xu
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
- *Correspondence: Yanmin Xu, ; Cheng Qian,
| | - Cheng Qian
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Yanmin Xu, ; Cheng Qian,
| |
Collapse
|
13
|
Eng C, Ciombor KK, Cho M, Dorth JA, Rajdev LN, Horowitz DP, Gollub MJ, Jácome AA, Lockney NA, Muldoon RL, Washington MK, O'Brian BA, Benny A, Lebeck Lee CM, Benson AB, Goodman KA, Morris VK. Anal Cancer: Emerging Standards in a Rare Rare Disease. J Clin Oncol 2022; 40:2774-2788. [PMID: 35649196 DOI: 10.1200/jco.21.02566] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The social stigma surrounding an anal cancer diagnosis has traditionally prevented open discussions about this disease. However, as recent treatment options and an increasing rate of diagnoses are made worldwide, awareness is growing. In the United States alone, 9,090 individuals were expected to be diagnosed with anal cancer in 2021. The US annual incidence of squamous cell carcinoma of the anus continues to increase by 2.7% yearly, whereas the mortality rate increases by 3.1%. The main risk factor for anal cancer is a human papillomavirus infection; those with chronic immunosuppression are also at risk. Patients with HIV are 19 times more likely to develop anal cancer compared with the general population. In this review, we have provided an overview of the carcinoma of the anal canal, the role of screening, advancements in radiation therapy, and current trials investigating acute and chronic treatment-related toxicities. This article is a comprehensive approach to presenting the existing data in an effort to encourage continuous international interest in anal cancer.
Collapse
Affiliation(s)
- Cathy Eng
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Kristen K Ciombor
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - May Cho
- Division of Hematology and Oncology, Department of Medicine, University of California- Irvine School of Medicine, Irvine, CA
| | - Jennifer A Dorth
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Lakshmi N Rajdev
- Division for Hematology and Oncology, Department of Medicine, Northwell Health/Lenox Hill Hospital, New York, NY
| | - David P Horowitz
- Department of Radiation Oncology, New York Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY
| | - Marc J Gollub
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Alexandre A Jácome
- OncoBio Comprehensive Cancer Center, Department of Gastrointestinal Medical Oncology, Nova Lima, Brazil
| | - Natalie A Lockney
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Roberta L Muldoon
- Division of Colon and Rectal Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Mary Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Brittany A O'Brian
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Amala Benny
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Cody M Lebeck Lee
- VA Tennessee Valley Healthcare System, Department of Internal Medicine, Nashville, TN
| | - Al B Benson
- Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Van Karlyle Morris
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
14
|
Trujillo-Cirilo L, Torres-Corioriles EI, Rangel-Corona R, Corona-Ortega MT, Weiss-Steider B. Evidence that the viral oncoproteins E6 and E7 of HPV induce the expression of a functional IL-2R on cervical cancer cells. Cytokine 2021; 148:155592. [DOI: https:/doi.org/10.1016/j.cyto.2021.155592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
|
15
|
Mbuya W, Held K, Mcharo RD, Haule A, Mhizde J, Mnkai J, Mahenge A, Mwakatima M, Sembo M, Mwalongo W, Agrea P, Hoelscher M, Maboko L, Saathoff E, Geisenberger O, Rwegoshora F, Torres L, Koup RA, Kroidl A, Chachage M, Geldmacher C. Depletion of Human Papilloma Virus E6- and E7-Oncoprotein-Specific T-Cell Responses in Women Living With HIV. Front Immunol 2021; 12:742861. [PMID: 34759925 PMCID: PMC8573218 DOI: 10.3389/fimmu.2021.742861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background Cervical cancer - caused by persistent High Risk Human Papilloma Virus (HR HPV) infections - is the second most common cancer affecting women globally. HIV infection increases the risk for HPV persistence, associated disease progression and malignant cell transformation. We therefore hypothesized that this risk increase is directly linked to HIV infection associated dysfunction or depletion of HPV-oncoprotein-specific T-cell responses. Methods The 2H study specifically included HIV+ and HIV- women with and without cervical lesions and cancer to analyze HPV oncogene-specific T cell responses in relation to HPV infection, cervical lesion status and HIV status. Oncoprotein E6 and E7 specific T-cell responses were quantified for the most relevant types HPV16, 18 and 45 and control antigens (CMV-pp65) and M.tb-PPD in 373 women, using fresh peripheral blood mononuclear cells in an IFN-γ release ELISpot assay. Results Overall, systemic E6- and E7-oncoprotein-specific T-cell responses were infrequent and of low magnitude, when compared to CMV-pp65 and M.tb-PPD (p < 0.001 for all HR HPV types). Within HIV negative women infected with either HPV16, 18 or 45, HPV16 infected women had lowest frequency of autologous-type-E6/E7-specific T-cell responses (33%, 16/49), as compared to HPV18 (46% (6/13), p = 0.516) and HPV45 (69% (9/13), p = 0.026) infected women. Prevalent HPV18 and 45, but not HPV16 infections were linked to detectable oncoprotein-specific T-cell responses, and for these infections, HIV infection significantly diminished T-cell responses targeting the autologous infecting genotype. Within women living with HIV, low CD4 T-cell counts, detectable HIV viremia as well as cancerous and precancerous lesions were significantly associated with depletion of HPV oncoprotein-specific T-cell responses. Discussion Depletion of HPV-oncoprotein-specific T-cell responses likely contributes to the increased risk for HR HPV persistence and associated cancerogenesis in women living with HIV. The low inherent immunogenicity of HPV16 oncoproteins may contribute to the exceptional potential for cancerogenesis associated with HPV16 infections.
Collapse
Affiliation(s)
- Wilbert Mbuya
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania.,Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ruby D Mcharo
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Antelmo Haule
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Jacklina Mhizde
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Jonathan Mnkai
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Anifrid Mahenge
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Maria Mwakatima
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Margareth Sembo
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Wolfram Mwalongo
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Peter Agrea
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Leonard Maboko
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania.,Tanzania Commission for AIDS (TACAIDS), Dar es Salaam, Tanzania
| | - Elmar Saathoff
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Otto Geisenberger
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - France Rwegoshora
- Pathology Department, Mbeya Zonal Referral Hospital, Mbeya, Tanzania
| | - Liset Torres
- Pathology Department, Mbeya Zonal Referral Hospital, Mbeya, Tanzania
| | - Richard A Koup
- Vaccine Research Centre, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Arne Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Mkunde Chachage
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania.,Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,Microbiology and Immunology Department, University of Dar es Salaam -Mbeya College of Health and Allied Sciences (UDSM-MCHAS), Mbeya, Tanzania
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| |
Collapse
|
16
|
Moniz CMV, Riechelmann RP, Oliveira SCR, Bariani GM, Rivelli TG, Ortega C, Pereira AAL, Meireles SI, Franco R, Chen A, Bonadio RC, Nahas C, Sabbaga J, Coudry RA, Braghiroli MI, Hoff PM. A Prospective Cohort Study of Biomarkers in Squamous Cell Carcinoma of the Anal Canal (SCCAC) and their Influence on Treatment Outcomes. J Cancer 2021; 12:7018-7025. [PMID: 34729104 PMCID: PMC8558650 DOI: 10.7150/jca.57678] [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: 12/29/2020] [Accepted: 10/05/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Although Chemoradiation (CRT) is the curative treatment for SCCAC, many patients present primary resistance. Since it is a rare tumor, response predictors remain unknown. Methods: We performed a prospective cohort study to evaluate biomarkers associated with CRT response, progression-free survival (PFS), and overall survival (OS). The primary endpoint was response at 6 months (m). Tumor DNA and HPV were analyzed by next-generation sequencing, while KI-67 and PD-L1 by immunohistochemistry in tumor tissue. Results: Seventy-eight patients were recruited between October/2011 and December/2015, and 75 were response evaluable. The median age was 57 years, 65% (n=49) were stage III and 12% (n=9) were HIV positive (HIV+). At 6m, 62.7% (n=47) presented CR. On multivariate analyses, stage II patients were 4.7 more likely to achieve response than stage III (OR, 4.70; 95%CI, 1.36-16.30; p=0.015). HIV+ was associated with a worse response (OR, 5.72; 95%CI, 2.5-13.0; p<0.001). 5-year PFS and OS rates were 63.3% and 76.4%, respectively, with a median follow up of 66m. On multivariate analyses, older age (HR 1.06, p=0.022, 95%IC 1.01-1.11) and absence of CR at 6m (HR 3.36, p=0.007, 95%IC 1.39-8.09) were associated with inferior OS. The 5-year OS rate was 62.5% in HIV+ group compared to 78% among HIV- pts, although this difference was not statistically significant (p=0.4). PIK3CA, MET and TP53 mutations, HPV, Ki-67 expression, and PD-L1 expression, were not associated with PFS and OS. Conclusions: Clinical stage III and HIV+ were associated with worse response to CRT at 6m. The absence of CR was the main factor associated with poor 5-year OS.
Collapse
Affiliation(s)
- Camila Motta Venchiarutti Moniz
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| | | | | | - Giovanni Mendonça Bariani
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Thomas Giollo Rivelli
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Cintia Ortega
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | | | - Rejane Franco
- Universidade Federal do Paraná - Hospital de Clínicas, Curitiba, PR, Brasil
| | - Andre Chen
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Renata Colombo Bonadio
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Caio Nahas
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Jorge Sabbaga
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Maria Ignez Braghiroli
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| | - Paulo Marcelo Hoff
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR.,Instituto D'Or de Pesquisa e Ensino - IDOR, Sao Paulo, SP, BR
| |
Collapse
|
17
|
Neckermann P, Boilesen DR, Willert T, Pertl C, Schrödel S, Thirion C, Asbach B, Holst PJ, Wagner R. Design and Immunological Validation of Macaca fascicularis Papillomavirus Type 3 Based Vaccine Candidates in Outbred Mice: Basis for Future Testing of a Therapeutic Papillomavirus Vaccine in NHPs. Front Immunol 2021; 12:761214. [PMID: 34777375 PMCID: PMC8581358 DOI: 10.3389/fimmu.2021.761214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/05/2021] [Indexed: 01/18/2023] Open
Abstract
Persistent human papillomavirus (HPV) infections are causative for cervical neoplasia and carcinomas. Despite the availability of prophylactic vaccines, morbidity and mortality induced by HPV are still too high. Thus, an efficient therapy, such as a therapeutic vaccine, is urgently required. Herein, we describe the development and validation of Macaca fascicularis papillomavirus type 3 (MfPV3) antigens delivered via nucleic-acid and adenoviral vectors in outbred mouse models. Ten artificially fused polypeptides comprising early viral regulatory proteins were designed and optionally linked to the T cell adjuvant MHC-II-associated invariant chain. Transfected HEK293 cells and A549 cells transduced with recombinant adenoviruses expressing the same panel of artificial antigens proved proper and comparable expression, respectively. Immunization of outbred CD1 and OF1 mice led to CD8+ and CD4+ T cell responses against MfPV3 antigens after DNA- and adenoviral vector delivery. Moreover, in vivo cytotoxicity of vaccine-induced CD8+ T cells was demonstrated in BALB/c mice by quantifying specific killing of transferred peptide-pulsed syngeneic target cells. The use of the invariant chain as T cell adjuvant enhanced the T cell responses regarding cytotoxicity and in vitro analysis suggested an accelerated turnover of the antigens as causative. Notably, the fusion-polypeptide elicited the same level of T-cell responses as administration of the antigens individually, suggesting no loss of immunogenicity by fusing multiple proteins in one vaccine construct. These data support further development of the vaccine candidates in a follow up efficacy study in persistently infected Macaca fascicularis monkeys to assess their potential to eliminate pre-malignant papillomavirus infections, eventually instructing the design of an analogous therapeutic HPV vaccine.
Collapse
Affiliation(s)
- Patrick Neckermann
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Ditte Rahbaek Boilesen
- Centre for Medical Parasitology, the Panum Institute, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- InProTher APS, Copenhagen, Denmark
| | | | | | | | | | - Benedikt Asbach
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Peter Johannes Holst
- Centre for Medical Parasitology, the Panum Institute, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- InProTher APS, Copenhagen, Denmark
| | - Ralf Wagner
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
18
|
Abstract
Owing to the presence of known tumor-specific viral antigens, human papillomavirus (HPV)-associated cancers are well suited for treatment with immunotherapy designed to unleash, amplify or replace the T cell arm of the adaptive immune system. Immune checkpoint blockade designed to unleash existing T cell immunity is currently Food and Drug Administration approved for certain HPV-associated cancers. More specific immunotherapies such as therapeutic vaccines and T cell receptor-engineered cellular therapy are currently in clinical development. Such therapies may offer more specific immune activation against viral tumor antigens and decrease the risk of immune-related adverse events. Current and planned clinical study of these treatments will determine their utility in the treatment of patients with newly diagnosed advanced stage or relapsed HPV-associated cancer.
Collapse
Affiliation(s)
- Maxwell Y Lee
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
19
|
Verdegaal E, van der Kooij MK, Visser M, van der Minne C, de Bruin L, Meij P, Terwisscha van Scheltinga A, Welters MJ, Santegoets S, de Miranda N, Roozen I, Liefers GJ, Kapiteijn E, van der Burg SH. Low-dose interferon-alpha preconditioning and adoptive cell therapy in patients with metastatic melanoma refractory to standard (immune) therapies: a phase I/II study. J Immunother Cancer 2021; 8:jitc-2019-000166. [PMID: 32238469 PMCID: PMC7174065 DOI: 10.1136/jitc-2019-000166] [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] [Accepted: 02/17/2020] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Adoptive cell therapy (ACT) with tumor-reactive T cells has shown consistent clinical efficacy. We evaluated the response to ACT in combination with interferon alpha (IFNa) preconditioning in patients with stage IV metastatic melanoma, most of which were progressive on cytotoxic T-lymphocyte-associated protein 4 and/or programmed cell death protein 1 checkpoint blockade therapy. METHODS Thirty-four patients were treated with ex vivo expanded tumor reactive T cells, derived from mixed lymphocyte autologous tumor cultures, or with autologous tumor-infiltrating lymphocytes and evaluated for clinical response. Clinical and immunological parameters associated with response were also evaluated. RESULTS Best overall response defined as clinical benefit, comprising either complete response, partial response or stable disease >6 months, was observed in 29% of the patients. Forty-three per cent of the 14 immunotherapy-naïve patients and 20% of the 20 patients progressive on prior immunotherapy benefited from ACT. The overall survival (OS) was 90% versus 28.6% at 1 year and 46.7% versus 0% at 3 years follow-up, of responder and non-responder patients, respectively. Median OS was 36 versus 7 months, respectively. IFNa pretreatment resulted in leukopenia, neutropenia and lymphopenia, which was sustained during the treatment in clinical responders and associated with response. Differences in antigen specificity, but not in phenotype, cytokine profile or CD8+ T cell number of the ACT products correlated with clinical response. Cross-reactivity of the ACT products to one or more allogeneic human leukocyte antigen-matched melanoma cell lines was associated with short OS after treatment while the ACT products of very long-term survivors showed no cross-reactivity but recognized patient-specific neoantigens. CONCLUSION This study demonstrates that ACT in combination with a mild IFNa preconditioning regimen can induce clinical benefit even in immunotherapy pretreated patients, although with lower success than in immunotherapy-naïve patients. ACT products comprising neoantigen reactivity may be more effective.
Collapse
Affiliation(s)
- Els Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique K van der Kooij
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Marten Visser
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Caroline van der Minne
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda de Bruin
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Pauline Meij
- GMP Facility Leiden, Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anton Terwisscha van Scheltinga
- GMP Facility Leiden, Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J Welters
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Saskia Santegoets
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Noel de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge Roozen
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerrit Jan Liefers
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Shamseddine AA, Burman B, Lee NY, Zamarin D, Riaz N. Tumor Immunity and Immunotherapy for HPV-Related Cancers. Cancer Discov 2021; 11:1896-1912. [PMID: 33990345 PMCID: PMC8338882 DOI: 10.1158/2159-8290.cd-20-1760] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Human papillomavirus (HPV) infection drives tumorigenesis in the majority of cervical, oropharyngeal, anal, and vulvar cancers. Genetic and epidemiologic evidence has highlighted the role of immunosuppression in the oncogenesis of HPV-related malignancies. Here we review how HPV modulates the immune microenvironment and subsequent therapeutic implications. We describe the landscape of immunotherapies for these cancers with a focus on findings from early-phase studies exploring antigen-specific treatments, and discuss future directions. Although responses across these studies have been modest to date, a deeper understanding of HPV-related tumor biology and immunology may prove instrumental for the development of more efficacious immunotherapeutic approaches. SIGNIFICANCE: HPV modulates the microenvironment to create a protumorigenic state of immune suppression and evasion. Our understanding of these mechanisms has led to the development of immunomodulatory treatments that have shown early clinical promise in patients with HPV-related malignancies. This review summarizes our current understanding of the interactions of HPV and its microenvironment and provides insight into the progress and challenges of developing immunotherapies for HPV-related malignancies.
Collapse
Affiliation(s)
- Achraf A Shamseddine
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bharat Burman
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dmitriy Zamarin
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
21
|
Al Bitar S, Ballouz T, Doughan S, Gali-Muhtasib H, Rizk N. Potential role of micro ribonucleic acids in screening for anal cancer in human papilloma virus and human immunodeficiency virus related malignancies. World J Gastrointest Pathophysiol 2021; 12:59-83. [PMID: 34354849 PMCID: PMC8316837 DOI: 10.4291/wjgp.v12.i4.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/24/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Despite advances in antiretroviral treatment (ART), human immunodeficiency virus (HIV) continues to be a major global public health issue owing to the increased mortality rates related to the prevalent oncogenic viruses among people living with HIV (PLWH). Human papillomavirus (HPV) is the most common sexually transmitted viral disease in both men and women worldwide. High-risk or oncogenic HPV types are associated with the development of HPV-related malignancies, including cervical, penile, and anal cancer, in addition to oral cancers. The incidence of anal squamous cell cancers is increasing among PLWH, necessitating the need for reliable screening methods in this population at risk. In fact, the currently used screening methods, including the Pap smear, are invasive and are neither sensitive nor specific. Investigators are interested in circulatory and tissue micro ribonucleic acids (miRNAs), as these small non-coding RNAs are ideal biomarkers for early detection and prognosis of cancer. Multiple miRNAs are deregulated during HIV and HPV infection and their deregulation contributes to the pathogenesis of disease. Here, we will review the molecular basis of HIV and HPV co-infections and focus on the pathogenesis and epidemiology of anal cancer in PLWH. The limitations of screening for anal cancer and the need for a reliable screening program that involves specific miRNAs with diagnostic and therapeutic values is also discussed.
Collapse
Affiliation(s)
- Samar Al Bitar
- Department of Biology, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Tala Ballouz
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Samer Doughan
- Department of Surgery, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Hala Gali-Muhtasib
- Department of Biology and Center for Drug Discovery, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Nesrine Rizk
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| |
Collapse
|
22
|
Melief CJM, Welters MJP, Vergote I, Kroep JR, Kenter GG, Ottevanger PB, Tjalma WAA, Denys H, van Poelgeest MIE, Nijman HW, Reyners AKL, Velu T, Goffin F, Lalisang RI, Loof NM, Boekestijn S, Krebber WJ, Hooftman L, Visscher S, Blumenstein BA, Stead RB, Gerritsen W, van der Burg SH. Strong vaccine responses during chemotherapy are associated with prolonged cancer survival. Sci Transl Med 2021; 12:12/535/eaaz8235. [PMID: 32188726 DOI: 10.1126/scitranslmed.aaz8235] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/18/2020] [Indexed: 12/23/2022]
Abstract
Therapeutic cancer vaccines have effectively induced durable regressions of premalignant oncogenic human papilloma virus type 16 (HPV16)-induced anogenital lesions. However, the treatment of HPV16-induced cancers requires appropriate countermeasures to overcome cancer-induced immune suppression. We previously showed that standard-of-care carboplatin/paclitaxel chemotherapy can reduce abnormally high numbers of immunosuppressive myeloid cells in patients, allowing the development of much stronger therapeutic HPV16 vaccine (ISA101)-induced tumor immunity. We now show the clinical effects of ISA101 vaccination during chemotherapy in 77 patients with advanced, recurrent, or metastatic cervical cancer in a dose assessment study of ISA101. Tumor regressions were observed in 43% of 72 evaluable patients. The depletion of myeloid suppressive cells by carboplatin/paclitaxel was associated with detection of low frequency of spontaneous HPV16-specific immunity in 21 of 62 tested patients. Patients mounted type 1 T cell responses to the vaccine across all doses. The group of patients with higher than median vaccine-induced immune responses lived longer, with a flat tail on the survival curve. This demonstrates that chemoimmunotherapy can be exploited to the benefit of patients with advanced cancer based on a defined mode of action.
Collapse
Affiliation(s)
- Cornelis J M Melief
- ISA Pharmaceuticals, J.H. Oortweg 19, 2333 CH Leiden, Netherlands. .,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Marij J P Welters
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Ignace Vergote
- Department of Gynecologic Oncology, University Hospital, Leuven Cancer Institute, UZ Herestraat 49, 3000 Leuven, Belgium
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Gemma G Kenter
- Center for Gynecologic Oncology Amsterdam, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Petronella B Ottevanger
- Department of Medical Oncology, Nijmegen University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, Netherlands
| | - Wiebren A A Tjalma
- Multidisciplinary Breast Clinic-Unit Gynecological Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Hannelore Denys
- Department of Medical Oncology, University Hospital, De Pintelaan 185, 9000 Gent, Belgium
| | | | - Hans W Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Anna K L Reyners
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Thierry Velu
- Chirec Cancer Institute, Medical Centre Edith Cavell, Rue Edith Cavell 32, 1180 Brussels, Belgium
| | - Frederic Goffin
- Chirec Cancer Institute, Medical Centre Edith Cavell, Rue Edith Cavell 32, 1180 Brussels, Belgium
| | - Roy I Lalisang
- Department of Medical Oncology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, Netherlands
| | - Nikki M Loof
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Sanne Boekestijn
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | | | - Leon Hooftman
- ISA Pharmaceuticals, J.H. Oortweg 19, 2333 CH Leiden, Netherlands
| | - Sonja Visscher
- ISA Pharmaceuticals, J.H. Oortweg 19, 2333 CH Leiden, Netherlands
| | | | - Richard B Stead
- BioPharma Consulting Services, 691 96th Avenue Southeast, Bellevue, WA 98004, USA
| | - Winald Gerritsen
- Department of Medical Oncology, Nijmegen University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, Netherlands
| | - Sjoerd H van der Burg
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, Netherlands. .,Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| |
Collapse
|
23
|
Trujillo-Cirilo L, Torres-Corioriles EI, Rangel-Corona R, Corona-Ortega MT, Weiss-Steider B. Evidence that the viral oncoproteins E6 and E7 of HPV induce the expression of a functional IL-2R on cervical cancer cells. Cytokine 2021; 148:155592. [PMID: 34099345 DOI: 10.1016/j.cyto.2021.155592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 11/19/2022]
Abstract
HPV-positive (HPV+) cervical cancer (CC) cells have been reported to express the IL-2 receptor (IL-2R) in contrast to virus-negative CC cells. This work was carried out to evaluate whether HPV infection induces IL-2R expression in CC cells. The analysis of the IL-2R expression data collected from The Cancer Genome Atlas (TCGA) and Genotype Tissue Expression project (GTEx) using the Xena platform demonstrate a higher expression of IL-2R subunits in CC tumors in comparison with normal tissues. Moreover IL-2Rβ expression is consistently higher in HPV+ tumors versus HPV- tumors. Furthermore, it was demonstrated that transfection of the HPV E6/E7 genes into the C33A (HPV-) cell line promotes IL-2R expression and regulates proliferation in response to exogenous IL-2. Additionally, we found that HPV+ cell lines enhances their proliferation in co-culture with peripheral blood lymphocytes (PBLs). To corroborate that the viral proteins E6 and E7 were related to the effects mediated by IL-2, we used cells derived from the HeLa cell line in which the expression of E6/E7 has decreased, we found that it loses the ability to respond to the exogenous IL-2 stimuli. Finally, the importance of IL-2R in CC, as an immune escape mechanism, is discussed.
Collapse
Affiliation(s)
- Leonardo Trujillo-Cirilo
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico.
| | - Edgar Ivan Torres-Corioriles
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico.
| | - Rosalva Rangel-Corona
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico.
| | - Maria Teresa Corona-Ortega
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico.
| | - Benny Weiss-Steider
- Laboratory of Cellular Oncology, Research Unit Cell Differentiation and Cancer, L-4 P.B. FES Zaragoza, National University of Mexico, Av., Guelatao No. 66 Col. Ejercito de Oriente, Iztapalapa, C.P. 09230 Mexico City, Mexico.
| |
Collapse
|
24
|
Eastern Canadian Gastrointestinal Cancer Consensus Conference 2019. ACTA ACUST UNITED AC 2021; 28:1988-2006. [PMID: 34073199 PMCID: PMC8161825 DOI: 10.3390/curroncol28030185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 01/06/2023]
Abstract
The annual Eastern Canadian Gastrointestinal Cancer Consensus Conference 2019 was held in Morell, Prince Edward Island, 19-21 September 2019. Experts in medical oncology, radiation oncology, and surgical oncology who are involved in the management of patients with gastrointestinal malignancies participated in presentations and discussion sessions for the purpose of developing the recommendations presented here. This consensus statement addresses multiple topics in the management of anal, colorectal, biliary tract, and gastric cancers, including: radiotherapy and systemic therapy for localized and advanced anal cancer; watch and wait strategy for the management of rectal cancer; role of testing for dihydropyrimidine dehydrogenase (DPD) deficiency prior to commencement of fluoropyrimidine therapy; radiotherapy and systemic therapy in the adjuvant and unresectable settings for biliary tract cancer; and radiotherapy and systemic therapy in the perioperative setting for early-stage gastric cancer.
Collapse
|
25
|
Fernandes ATG, Carvalho MOO, Avvad-Portari E, Rocha NP, Russomano F, Roma EH, Bonecini-Almeida MDG. A prognostic value of CD45RA +, CD45RO +, CCL20 + and CCR6 + expressing cells as 'immunoscore' to predict cervical cancer induced by HPV. Sci Rep 2021; 11:8782. [PMID: 33888832 PMCID: PMC8062468 DOI: 10.1038/s41598-021-88248-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
The interplay between cervical cancer (CC) and immune cells, mainly intratumoral lymphocytes, has a pivotal role in carcinogenesis. In this context, we evaluated the distribution of CD45RA+ and CD45RO+ cells as well as CCR6+ and CCL20+ cells in intraepithelial (IE) and marginal stroma (MS) areas from cervical intraepithelial neoplasia (CIN) I-III, and CC as 'immunoscore' for HPV-induced CC outcome. We observed increased CD45RA+ and CD45RO+ cells distribution in IE and MS areas in the CC group compared to CIN groups and healthy volunteers. Interestingly, there is a remarkable reduction of CCL20+ expressing cells distribution according to lesion severity. The CC group had a significant decrease in CCL20+ and CCR6+-expressing cells distribution in both IE and MS areas compared to all groups. Using the 'immunoscore' model, we observed an increased number of women presenting high CD45RA+/CD45RO+ and low CCL20+/CCR6+ 'immunoscore' in the CC group. Our results suggested a pattern in cervical inflammatory process with increasing CD45RA+/CD45RO+, and decreasing CCL20+/CCR6+ expression in accordance with CIN severity. Taken together, these markers could be evaluated as 'immunoscore' predictors to CC response. A more comprehensive analysis of longitudinal studies should be conducted to associate CD45RA+/CD45RO+ and CCL20+/CCR6+ 'immunoscore' to CC progression and validate its value as a prognosis method.
Collapse
Affiliation(s)
- Ana Teresa G Fernandes
- Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil.
| | - Maria Odete O Carvalho
- Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Elyzabeth Avvad-Portari
- Department of Pathologic Anatomy at Fernandes Figueira Woman, Child and Adolescent's Health National Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Natália P Rocha
- Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Fabio Russomano
- Department of Gynecology at Fernandes Figueira Woman, Child and Adolescent's Health National Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Eric Henrique Roma
- Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Maria da Gloria Bonecini-Almeida
- Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| |
Collapse
|
26
|
Richardson JR, Schöllhorn A, Gouttefangeas C, Schuhmacher J. CD4+ T Cells: Multitasking Cells in the Duty of Cancer Immunotherapy. Cancers (Basel) 2021; 13:596. [PMID: 33546283 PMCID: PMC7913359 DOI: 10.3390/cancers13040596] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer immunotherapy activates the immune system to specifically target malignant cells. Research has often focused on CD8+ cytotoxic T cells, as those have the capacity to eliminate tumor cells after specific recognition upon TCR-MHC class I interaction. However, CD4+ T cells have gained attention in the field, as they are not only essential to promote help to CD8+ T cells, but are also able to kill tumor cells directly (via MHC-class II dependent recognition) or indirectly (e.g., via the activation of other immune cells like macrophages). Therefore, immunotherapy approaches have shifted from only stimulating CD8+ T cells to targeting and assessing both, CD4+ and CD8+ T cell subsets. Here, we discuss the various subsets of CD4+ T cells, their plasticity and functionality, their relevance in the antitumor immune response in patients affected by cancer, and their ever-growing role in therapeutic approaches for human cancer.
Collapse
Affiliation(s)
- Jennifer R. Richardson
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
| | - Anna Schöllhorn
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany
| | - Juliane Schuhmacher
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
27
|
Litwin TR, Irvin SR, Chornock RL, Sahasrabuddhe VV, Stanley M, Wentzensen N. Infiltrating T-cell markers in cervical carcinogenesis: a systematic review and meta-analysis. Br J Cancer 2021; 124:831-841. [PMID: 33257839 PMCID: PMC7884592 DOI: 10.1038/s41416-020-01184-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The host adaptive immune response helps determine which cervical HPV infections persist and progress to precancer and cancer, and systematic characterisation of T-cell infiltration would help inform key steps in cervical carcinogenesis. METHODS A systematic review and meta-analysis were conducted of infiltrating T-cells in normal cervix, low-grade lesions, high-grade lesions, and invasive cancers including epithelial, stromal, and total tissue and the following markers: CD3, CD4, CD8, FoxP3, CD25, and the CD4:CD8 ratio. An additional qualitative review summarised longitudinal data on associations between infiltrating T-cells and cervical disease persistence, regression, progression, or prognosis. RESULTS There were fewer CD3+, CD4+, and CD8+ cells in cervical lesions and more cells in cancers compared to normal epithelium. FoxP3 and CD25+ regulatory T-cell infiltration is high in persistent and precancerous lesions, and longitudinal data show improved outcomes with lower regulatory T-cell levels. CONCLUSIONS Successful immune evasion may reduce T-cell infiltration in HPV infected and precancerous epithelium, while invasive cancers are highly immunogenic, and regulatory T-cell infiltration increases with cervical disease progression. Understanding these factors may have prognostic value and could aid in novel treatment development and clinical guidelines, but published data are highly heterogeneous and leave important gaps to be filled by future studies.
Collapse
Affiliation(s)
- Tamara R Litwin
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| | - Sarah R Irvin
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rebecca L Chornock
- Department of Obstetrics and Gynecology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Vikrant V Sahasrabuddhe
- Breast and Gynecologic Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | | | - Nicolas Wentzensen
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
28
|
Abstract
In persistent high-risk HPV infection, viral gene expression can trigger some important early changes to immune capabilities which act to protect the lesion from immune attack and subsequently promote its growth and ability for sustained immune escape. This includes immune checkpoint-inhibitor ligand expression (e.g. PD-L1) by tumour or associated immune cells that can block any anti-tumour T-cell effectors. While there are encouraging signs of efficacy for cancer immunotherapies including with immune checkpoint inhibitors, therapeutic vaccines and adoptive cell therapies, overall response and survival rates remain relatively low. HPV oncogene vaccination has shown some useful efficacy in treatment of patients with high-grade lesions but was unable to control later stage cancers. To maximally exploit anti-tumour immune responses, the suppressive factors associated with HPV carcinogenesis must be countered. Importantly, a combination of chemotherapy, reducing immunosuppressive myeloid cells, with therapeutic HPV vaccination significantly improves impact on cancer treatment. Many clinical trials are investigating checkpoint inhibitor treatments in HPV associated cancers but response rates are limited; combination with vaccination is being tested. Further investigation of how chemo- and/or radio-therapy can influence the recovery of effective anti-tumour immunity is warranted. Understanding how to optimally deploy and sequence conventional and immunotherapies is the challenge.
Collapse
|
29
|
Paaso A, Koskimaa HM, Welters MJP, Kero K, Rautava J, Syrjänen K, van der Burg SH, Syrjänen S. Interferon-γ and IL-5 associated cell-mediated immune responses to HPV16 E2 and E6 distinguish between persistent oral HPV16 infections and noninfected mucosa. Clin Exp Dent Res 2021; 7:903-913. [PMID: 33421352 PMCID: PMC8543460 DOI: 10.1002/cre2.396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 10/27/2020] [Accepted: 12/23/2020] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Natural history of human papillomavirus (HPV) infection in the head and neck region is poorly understood, and their impact on collective HPV-specific immunity is not known. MATERIALS AND METHODS In this study, we have performed a systematic analysis of HPV16-specific cell-mediated immunity (CMI) in 21 women with known oral and genital HPV DNA status and HPV serology (Ab) based on 6-year follow-up data. These women being a subgroup from the Finnish Family HPV Study were recalled for blood sampling to be tested for their CMI-responses to HPV16 E2, E6, and E7 peptides. RESULTS The results showed that HPV16 E2-specific lymphocyte proliferation was more prevalent in women who tested HPV16 DNA negative in oral mucosa and were either HPV16 seropositive or negative than in HPV16 DNA+/Ab+ women (p = 0.046 and p = 0.035). In addition, the HPV16 DNA-/Ab- women most often displayed E6-specific proliferation (p = 0.020). Proportional cytokine profiles indicated that oral HPV16-negative women were characterized by prominent IFN-γ and IL-5 secretion not found in women with persisting oral HPV16 (p = 0.014 and p = 0.040, respectively). CONCLUSIONS Our results indicate that the naturally arising immune response induced by oral HPV infections displays a mixed Th1/Th2/Th17 cytokine profile while women with persisting oral HPV16 might have an impaired HPV16-specific CMI, shifted partly toward a Th2 profile, similarly as seen earlier among patients with high-grade genital HPV lesions. Thus, the lack of HPV 16 E2 and E6 specific T memory cells and Th2 cytokines might also predispose women for persistent oral HPV16 infection which might be related to the risk of cancer.
Collapse
Affiliation(s)
- Anna Paaso
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland
| | - Hanna-Mari Koskimaa
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland
| | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Katja Kero
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku, Finland
| | - Jaana Rautava
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland.,Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kari Syrjänen
- Department of Clinical Research, Biohit Oyj, Helsinki, Finland
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stina Syrjänen
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland.,Department of Pathology, Turku University Hospital, Turku, Finland
| |
Collapse
|
30
|
Welters MJP, Santegoets SJ, van der Burg SH. The Tumor Microenvironment and Immunotherapy of Oropharyngeal Squamous Cell Carcinoma. Front Oncol 2020; 10:545385. [PMID: 33425717 PMCID: PMC7793705 DOI: 10.3389/fonc.2020.545385] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022] Open
Abstract
Oropharyngeal squamous cell carcinoma (OPSCC) develops as a consequence of several mutations in the tumor suppressor pathways or after a progressive infection with high risk human papillomavirus (HPV). The dismal side effects of the current standard of care and the clear involvement of the immune system has led to a surge in clinical trials that aim to reinforce the tumor-specific immune response as a new treatment option. In this review, we have focused on the most recent literature to discuss the new findings and insights on the role of different immune cells in the context of OPSCC and its etiology. We then applied this knowledge to describe potential biomarkers and analyzed the rationale and outcomes of earlier and ongoing immunotherapy trials. Finally, we describe new developments that are still at the preclinical phase and provide an outlook on what the near future may bring, now that several new and exciting techniques to study the immune system at the single cell level are being exploited.
Collapse
Affiliation(s)
- Marij J P Welters
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Saskia J Santegoets
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Sjoerd H van der Burg
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
31
|
Komdeur FL, Singh A, van de Wall S, Meulenberg JJM, Boerma A, Hoogeboom BN, Paijens ST, Oyarce C, de Bruyn M, Schuuring E, Regts J, Marra R, Werner N, Sluis J, van der Zee AGJ, Wilschut JC, Allersma DP, van Zanten CJ, Kosterink JGW, Jorritsma-Smit A, Yigit R, Nijman HW, Daemen T. First-in-Human Phase I Clinical Trial of an SFV-Based RNA Replicon Cancer Vaccine against HPV-Induced Cancers. Mol Ther 2020; 29:611-625. [PMID: 33160073 PMCID: PMC7854293 DOI: 10.1016/j.ymthe.2020.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/08/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022] Open
Abstract
A first-in-human phase I trial of Vvax001, an alphavirus-based therapeutic cancer vaccine against human papillomavirus (HPV)-induced cancers was performed assessing immunological activity, safety, and tolerability. Vvax001 consists of replication-incompetent Semliki Forest virus replicon particles encoding HPV16-derived antigens E6 and E7. Twelve participants with a history of cervical intraepithelial neoplasia were included. Four cohorts of three participants were treated per dose level, ranging from 5 × 105 to 2.5 × 108 infectious particles per immunization. The participants received three immunizations with a 3-week interval. For immune monitoring, blood was drawn before immunization and 1 week after the second and third immunization. Immunization with Vvax001 was safe and well tolerated, with only mild injection site reactions, and resulted in both CD4+ and CD8+ T cell responses against E6 and E7 antigens. Even the lowest dose of 5 × 105 infectious particles elicited E6/E7-specific interferon (IFN)-γ responses in all three participants in this cohort. Overall, immunization resulted in positive vaccine-induced immune responses in 12 of 12 participants in one or more assays performed. In conclusion, Vvax001 was safe and induced immune responses in all participants. These data strongly support further clinical evaluation of Vvax001 as a therapeutic vaccine in patients with HPV-related malignancies.
Collapse
Affiliation(s)
- Fenne L Komdeur
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Amrita Singh
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Stephanie van de Wall
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Annemarie Boerma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Baukje Nynke Hoogeboom
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sterre T Paijens
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cesar Oyarce
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joke Regts
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ruben Marra
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Naomi Werner
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jessica Sluis
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ate G J van der Zee
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan C Wilschut
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Derk P Allersma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Coba J van Zanten
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jos G W Kosterink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annelies Jorritsma-Smit
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Refika Yigit
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Toos Daemen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
32
|
Yuan Y, Cai X, Shen F, Ma F. HPV post-infection microenvironment and cervical cancer. Cancer Lett 2020; 497:243-254. [PMID: 33122098 DOI: 10.1016/j.canlet.2020.10.034] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/12/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023]
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted virus worldwide. More than 99% of cervical cancer cases are associated with certain types of HPVs, termed high-risk types. In addition to the well-known transformative properties, HPVs-infected cells actively instruct the local milieu and create a supportive post-infection microenvironment (PIM), which is becoming recognized as a key factor for the viral persistence, propagation, and malignant progression. The PIM is initiated and established via a complex interplay among virus-infected cells, immune cells, and host stroma, as well as their derived components including chemokines, cytokines, extracellular vesicles, and metabolites. In this review, we summarize the current understanding of these key components, characteristics, and effects of the PIM, and highlights the prospect of targeting the PIM as a potential strategy to improve therapeutic outcomes for cervical cancer.
Collapse
Affiliation(s)
- Yi Yuan
- Suzhou Institute of Systems Medicine, Suzhou, 215123, China; Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine of Tongji University, Shanghai, 200065, China
| | - Xushan Cai
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Jiading District, Shanghai, 201821, China
| | - Fangrong Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, 215001, China.
| | - Feng Ma
- Suzhou Institute of Systems Medicine, Suzhou, 215123, China.
| |
Collapse
|
33
|
Coinfection of High-Risk Human Papillomavirus and Lower Genital Tract Pathogens in the Development of High-Grade Cervical Lesions. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2020; 2020:7640758. [PMID: 32908620 PMCID: PMC7477613 DOI: 10.1155/2020/7640758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/28/2020] [Accepted: 08/18/2020] [Indexed: 11/18/2022]
Abstract
Purpose This study investigated the infection status and relationship between other common lower genital tract infectious pathogens and high-risk human papillomavirus (HR-HPV) in the high-grade cervical lesions. Methods Overall, 882 patients were enrolled in this retrospective study, of which 339 patients (≥HSIL group) were confirmed with high-grade squamous intraepithelial lesions (HSIL) or cervical squamous cell carcinoma (SCC), while 543 patients (≤LSIL group) were diagnosed with low-grade squamous intraepithelial lesions (LSIL) or normal cervical pathology diagnosis. Cervical swab specimens were tested for HPV, pathogenic bacteria (PB), U. urealyticum (UU), M. hominis (MH), and C. trachomatis (CT) in both groups. Results The infection rates of HR-HPV, PB, UU (at high density), and CT were higher in the ≥HSIL group than in the ≤LSIL group (P < 0.001); however, higher infection rates with MH were not observed (P > 0.05). PB, UU, and CT were associated with HR-HPV infection (P < 0.001). The PB and UU infection rates in the ≥HSIL group were significantly different from those in the ≤LSIL group, regardless of whether there was an HR-HPV infection at the same time (P < 0.05). However, this was not the case for the CT (P > 0.05). Furthermore, 259 pathogenic bacterial strains were detected in 882 cases. The difference in the distribution of pathogenic bacterial flora in the different grades of cervical lesions had no statistical significance, which was prioritized over Escherichia coli (P > 0.05). Conclusion PB, UU, and CT infection is associated with susceptibility to HR-HPV, HR-HPV coinfection with these pathogens might increase the risk of high-grade cervical lesions, and PB and UU might be independent risk factors for cervical lesions.
Collapse
|
34
|
Pessia B, Romano L, Giuliani A, Lazzarin G, Carlei F, Schietroma M. Squamous cell anal cancer: Management and therapeutic options. Ann Med Surg (Lond) 2020; 55:36-46. [PMID: 32461801 PMCID: PMC7240186 DOI: 10.1016/j.amsu.2020.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/02/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023] Open
Abstract
The incidence of anal cancer has increased during the second half of the 20th century, with an incidence rate over 2.9% greater than in the decade of 1992-2001. Yet, it still constitutes a small percentage, about 4%, of all anorectal tumours. Its risk factors are human papillomavirus infection, a history of sexually transmitted diseases, a history of vulvar or cervical carcinoma, immunosuppression related to human immunodeficiency virus infection or after organ transplantation, haematological or immunological disorders, and smoking. The most frequent symptom is rectal bleeding (45%), followed by anal pain, and sensation of a rectal mass. The diagnosis requires clinical examination, palpation of the inguinal lymph nodes, high resolution anoscopy followed by fine-needle aspiration biopsy or core biopsy. Subsequent histologic diagnosis is necessary, as well as computed tomography or magnetic resonance imaging evaluation of the pelvic lymph nodes. Since 1980, patients with a diagnosis of anal cancer have shown a significant improvement in survival. In Europe during the years 1983-1994, 1-year survival increased from 78% to 81%, and the improvement over 5 years was between 48% and 54%. Prior to 1974, patients with invasive cancer were routinely scheduled for abdominoperineal amputation, after which it was demonstrated that treatment with 5-fluorouracil and radiotherapy associated with mitomycin or capecitabine could be adequate to treat the tumour without surgery. Today, numerous studies have confirmed that combined multimodal treatment is effective and sufficient.
Collapse
Affiliation(s)
- Beatrice Pessia
- Department of Surgery, Department of Applied Clinical Science and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Lucia Romano
- Department of Surgery, Department of Applied Clinical Science and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Antonio Giuliani
- Department of Surgery, Department of Applied Clinical Science and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Gianni Lazzarin
- Department of Surgery, Department of Applied Clinical Science and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Francesco Carlei
- Department of Surgery, Department of Applied Clinical Science and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Mario Schietroma
- Department of Surgery, Department of Applied Clinical Science and Biotechnology, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
35
|
Saint A, Evesque L, François É. [Metastatic squamous cell carcinomas of the anal canal: Current management and prospects]. Bull Cancer 2020; 107:792-799. [PMID: 32591138 DOI: 10.1016/j.bulcan.2020.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/18/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
Anal canal cancer is a rare disease that accounts for 2.5% of digestive cancers. Squamous cell carcinomas are the most common histological form. Their incidence is in progression, probably due to the increase in Human Papilloma Virus infections. Metastatic forms account for 20% of anal canal cancers considering synchronous forms or metastatic recurrence of an initially localised disease. Their prognosis remains poor with an estimated 5-year survival rate of 30%. The first-line therapeutic standard based on the combination of cisplatin with 5-Fluorouracil has recently been challenged by carboplatin - paclitaxel and docetaxel, cisplatin and 5-Fluorouracil regimens which are becoming new treatment options. In second-line setting, there is no international consensus. Anti-EGFRs and immunotherapy in combination or not with other molecules are promising but these results need to be confirmed. In this review, we report current and future data in the management of squamous cell carcinomas of the anal canal in unresectable locoregional recurrence or at metastatic stage.
Collapse
Affiliation(s)
- Angélique Saint
- Centre Antoine-Lacassagne, département d'oncologie médicale, Nice, France.
| | - Ludovic Evesque
- Centre Antoine-Lacassagne, département d'oncologie médicale, Nice, France
| | - Éric François
- Centre Antoine-Lacassagne, département d'oncologie médicale, Nice, France
| |
Collapse
|
36
|
Eng C, Fakih M, Amin M, Morris V, Hochster HS, Boland PM, Uronis H. A phase II study of axalimogene filolisbac for patients with previously treated, unresectable, persistent/recurrent loco-regional or metastatic anal cancer. Oncotarget 2020; 11:1334-1343. [PMID: 32341753 PMCID: PMC7170499 DOI: 10.18632/oncotarget.27536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/14/2020] [Indexed: 12/31/2022] Open
Abstract
Squamous cell carcinoma of the anorectal canal (SCCA) is a rare HPV-related malignancy that is steadily increasing in incidence. A high unmet need exists for patients with persistent loco-regional and metastatic disease. Axalimogene filolisbac (ADXS11-001) is an investigational immunotherapy that stimulates tumor-specific responses against HPV-associated cancers, and has demonstrated benefit in metastatic cervical cancer. We conducted this single-arm, multicenter, phase 2 trial in patients with persistent/recurrent, loco-regional or metastatic SCCA. Patients received ADXS11-001, 1 × 109 colony-forming units intravenously every 3 weeks. A Simon 2-stage design was used to test primary co-endpoints of overall response rate (ORR) and 6-month progression-free survival (PFS) rate. Study would proceed to full enrollment if ORR ≥ 10% or 6-month PFS rate ≥ 20%. Thirty-six patients were treated; 29 patients were evaluable for response. One patient had a prolonged partial response (3.4% ORR). The 6-month PFS rate was 15.5%. Grade 3 adverse event were noted in 10 patients, with the majority being cytokine-release symptoms; one grade 4 adverse event was noted. No grade 5 adverse events occurred. ADXS11-001 was safe and well-tolerated in patients with SCCA. However, this study did not meet either primary endpoint. ADXS11-001 may be more beneficial when administered in combination with other cytotoxic or targeted agents.
Collapse
Affiliation(s)
- Cathy Eng
- MD Anderson Cancer Center, Houston, TX, USA
| | | | - Manik Amin
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Van Morris
- MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | |
Collapse
|
37
|
Garbuglia AR, Lapa D, Sias C, Capobianchi MR, Del Porto P. The Use of Both Therapeutic and Prophylactic Vaccines in the Therapy of Papillomavirus Disease. Front Immunol 2020; 11:188. [PMID: 32133000 PMCID: PMC7040023 DOI: 10.3389/fimmu.2020.00188] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/24/2020] [Indexed: 12/30/2022] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted virus. The high-risk HPV types (i.e., HPV16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59) are considered to be the main etiological agents of genital tract cancers, such as cervical, vulvar, vaginal, penile, and anal cancers, and of a subset of head and neck cancers. Three prophylactic HPV vaccines are available that are bivalent (vs. HPV16, 18), tetravalent (vs. HPV6, 11, 16, 18), and non-avalent (vs. HPV6, 11, 16, 18, 31, 33,45, 52, 58). All of these vaccines are based on recombinant DNA technology, and they are prepared from the purified L1 protein that self-assembles to form the HPV type-specific empty shells (i.e., virus-like particles). These vaccines are highly immunogenic and induce specific antibodies. Therapeutic vaccines differ from prophylactic vaccines, as they are designed to generate cell-mediated immunity against transformed cells, rather than neutralizing antibodies. Among the HPV proteins, the E6 and E7 oncoproteins are considered almost ideal as targets for immunotherapy of cervical cancer, as they are essential for the onset and evolution of malignancy and are constitutively expressed in both premalignant and invasive lesions. Several strategies have been investigated for HPV therapeutic vaccines designed to enhance CD4+ and CD8+ T-cell responses, including genetic vaccines (i.e., DNA/ RNA/virus/ bacterial), and protein-based, peptide-based or dendritic-cell-based vaccines. However, no vaccine has yet been licensed for therapeutic use. Several studies have suggested that administration of prophylactic vaccines immediately after surgical treatment of CIN2 cervical lesions can be considered as an adjuvant to prevent reactivation or reinfection, and other studies have described the relevance of prophylactic vaccines in the management of genital warts. This review summarizes the leading features of therapeutic vaccines, which mainly target the early oncoproteins E6 and E7, and prophylactic vaccines, which are based on the L1 capsid protein. Through an analysis of the specific immunogenic properties of these two types of vaccines, we discuss why and how prophylactic vaccines can be effective in the treatment of HPV-related lesions and relapse.
Collapse
Affiliation(s)
- Anna Rosa Garbuglia
- Laboratory of Virology, "Lazzaro Spallanzani" National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Daniele Lapa
- Laboratory of Virology, "Lazzaro Spallanzani" National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Catia Sias
- Laboratory of Virology, "Lazzaro Spallanzani" National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, "Lazzaro Spallanzani" National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Paola Del Porto
- Department of Biology and Biotechnology "C. Darwin," Sapienza University, Rome, Italy
| |
Collapse
|
38
|
Wang R, Pan W, Jin L, Huang W, Li Y, Wu D, Gao C, Ma D, Liao S. Human papillomavirus vaccine against cervical cancer: Opportunity and challenge. Cancer Lett 2020; 471:88-102. [DOI: 10.1016/j.canlet.2019.11.039] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022]
|
39
|
Chambuso R, Ramesar R, Kaambo E, Murahwa AT, Abdallah MO, De Sousa M, Denny L, Williamson AL, Gray CM. Age, absolute CD4 count, and CD4 percentage in relation to HPV infection and the stage of cervical disease in HIV-1-positive women. Medicine (Baltimore) 2020; 99:e19273. [PMID: 32118737 PMCID: PMC7478573 DOI: 10.1097/md.0000000000019273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A subgroup of women who are co-infected with human immunodeficiency virus type 1 (HIV-1) and human papillomavirus (HPV), progress rapidly to cervical disease. We characterized HPV genotypes within cervical tumor biopsies, assessed the relationships of cervical disease stage with age, HIV-1 status, absolute CD4 count, and CD4 percentage, and identified the predictive power of these variables for cervical disease stage in a cohort of South African women.We recruited 181 women who were histologically diagnosed with cervical disease; 87 were HIV-1-positive and 94 were HIV-1-seronegative. Colposcopy-directed tumor biopsies were confirmed by histology and used for genomic DNA extraction. The Roche Linear Array HPV genotyping test was used for HPV genotyping. Peripheral whole blood was used for HIV-1 rapid testing. Fully automated FC500MPL/CellMek with PanLeucogate (PLG) was used to determine absolute CD4 count, CD4 percentage, and CD45 count. Chi-squared test, a logistic regression model, parametric Pearson correlation, and ROC curves were used for statistical analyses. We used the Benjamini-Horchberg test to control for false discovery rate (FDR, q-value). All tests were significant when both P and q were <.05.Age was a significant predictor for invasive cervical cancer (ICC) in both HIV-1-seronegative (P < .0001, q < 0.0001) and HIV-1-positive women (P = .0003, q = 0.0003). Sixty eight percent (59/87) of HIV-1-positive women with different stages of cervical disease presented with a CD4 percentage equal or less than 28%, and a median absolute CD4 count of 400 cells/μl (IQR 300-500 cells/μl). Of the HIV-1-positive women, 75% (30/40) with ICC, possessed ≤28% CD4 cells vs 25% (10/40) who possessed >28% CD4 cells (both P < .001, q < 0.001). Furthermore, 70% (28/40) of women with ICC possessed CD4 count >350 compared to 30% (12/40) who possessed CD4 count ≤ 350 (both P < .001, q < 0.001).Age is an independent predictor for ICC. In turn, development of ICC in HIV-1-positive women is independent of the host CD4 cells and associates with low CD4 percentage regardless of absolute CD4 count that falls within the normal range. Thus, using CD4 percentage may add a better prognostic indicator of cervical disease stage than absolute CD4 count alone.
Collapse
Affiliation(s)
- Ramadhani Chambuso
- MRC Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology
- Department of Gynaecology, Morogoro Regional Referral Hospital, Morogoro, Tanzania
| | - Raj Ramesar
- MRC Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology
- Division of Human Genetics
| | - Evelyn Kaambo
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences
- Department of Biochemistry and Medical Microbiology, University of Namibia School of Medicine, Windhoek, Namibia
| | | | - Mohammed O.E. Abdallah
- MRC Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology
- Department of Gynaecology, Morogoro Regional Referral Hospital, Morogoro, Tanzania
| | - Michelle De Sousa
- Department of Obstetrics and Gynaecology, Victoria Wynberg Hospital, Cape Town
- South African Medical Research Council, Clinical Gynaecological Cancer Research Centre
| | - Lynette Denny
- South African Medical Research Council, Clinical Gynaecological Cancer Research Centre
- Department of Obstetrics and Gynaecology
| | - Anna-Lise Williamson
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences
- South African Medical Research Council, Clinical Gynaecological Cancer Research Centre
| | - Clive M. Gray
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine and Department of Pathology, University of Cape Town
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
40
|
Liebenberg LJP, McKinnon LR, Yende-Zuma N, Garrett N, Baxter C, Kharsany ABM, Archary D, Rositch A, Samsunder N, Mansoor LE, Passmore JAS, Abdool Karim SS, Abdool Karim Q. HPV infection and the genital cytokine milieu in women at high risk of HIV acquisition. Nat Commun 2019; 10:5227. [PMID: 31745084 PMCID: PMC6863918 DOI: 10.1038/s41467-019-13089-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/18/2019] [Indexed: 01/05/2023] Open
Abstract
Human papillomavirus (HPV) infection correlates with higher rates of HIV acquisition, but the underlying biological mechanisms are unclear. Here we study associations between HPV and HIV acquisition and relate these to vaginal cytokine profiles in an observational cohort of women at high risk of HIV infection (CAPRISA 004, n = 779) and with 74% HPV prevalence. We report here that HPV infection associates with a 2.5-fold increase in HIV acquisition risk in this population (95% CI: 1.2-5.3). Among 48 vaginal cytokines profiled, cytokines associated with HPV infection overlap substantially with cytokines associated with HIV risk, but are distinct from those observed in HPV negative women. Although our data do not establish a causative link between HPV status and the risk of HIV, we suggest that increasing HPV vaccination coverage may carry an additional benefit of reducing the risk of contracting HIV infection, particularly in regions with high HPV prevalence.
Collapse
Affiliation(s)
- Lenine J P Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa.
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Nonhlanhla Yende-Zuma
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Cheryl Baxter
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Ayesha B M Kharsany
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Anne Rositch
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Natasha Samsunder
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Leila E Mansoor
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Jo-Ann S Passmore
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM) and MRC-UCT Gynecological Cancer Research Centre, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Epidemiology, Columbia University, New York City, NY, USA
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Epidemiology, Columbia University, New York City, NY, USA
| |
Collapse
|
41
|
Liu Y, Li H, Pi R, Yang Y, Zhao X, Qi X. Current strategies against persistent human papillomavirus infection (Review). Int J Oncol 2019; 55:570-584. [PMID: 31364734 DOI: 10.3892/ijo.2019.4847] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/16/2019] [Indexed: 11/06/2022] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted infection, exhibiting a tropism for the epidermis and mucosae. The link between persistent HPV infection and malignancies involving the anogenital tract as well as the head and neck has been well‑established, and it is estimated that HPV‑related cancers involving various anatomical sites account for 4.5% of all human cancers. Current prophylactic vaccines against HPV have enabled the prevention of associated malignancies. However, the sizeable population base of current infection in whom prophylactic vaccines are not applicable, certain high‑risk HPV types not included in vaccines, and the vast susceptible population in developing countries who do not have access to the costly prophylactic vaccines, put forward an imperative need for effective therapies targeting persistent infection. In this article, the life cycle of HPV, the mechanisms facilitating HPV evasion of recognition and clearance by the host immune system, and the promising therapeutic strategies currently under investigation, particularly antiviral drugs and therapeutic vaccines, are reviewed.
Collapse
Affiliation(s)
- Yu Liu
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ruyu Pi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
42
|
Ramos-Martínez E, Rojas-Serrano J, García-Hernández O, García-Vázquez FJ, Andrade WA, Avila G, Salinas-Pasquier L, López-Vancell MR. The immune response to Hymenolepis nana in mice decreases tumorigenesis induced by 7,12 dimethylbenz-anthracene. Cytokine 2019; 123:154743. [PMID: 31255915 DOI: 10.1016/j.cyto.2019.154743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/15/2019] [Accepted: 06/03/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cancer is a high-impact disease throughout the world. A negative correlation has been established between the development of cancer and the Th2 immune response. Infection by helminth parasites is characterized by the induction of a strong and long-lasting Th2 response. The aim of this work was to evaluate the effect of the immune response induced by the infection with the helminth Hymenolepis nana, on the tumorigenesis induced by dimethylbenz-anthracene (DMBA) in mice. METHODOLOGY Four different groups of 14 female BALB/c mice were formed; Group A, dimethyl sulfoxide (DMSO) (vehicle) was administered cutaneously, Group B infected with H. nana, group C, cutaneously DMBA and finally Group D infected with H. nana and cutaneous DMBA. The tumor load was determined in those animals that developed cancerous lesions. In all groups were determined: serum concentration of IgE, IFNγ, IL-10, IL-5 and malondialdehyde (MDA). The inflammatory infiltrate was analyzed from skin samples and the expression of the main eosinophilic protein and myeloperoxidase was determined. RESULTS The group previously infected with H. nana had a reduced amount of tumors with smaller size, in comparison to the group that received only DMBA; this reduction was associated with lower levels of IFNγ and IL-10, while levels of IgE, IL-5 and MDA were higher. Further, the number of eosinophils and neutrophils was statistically higher in the animals that were previously infected with the helminth and developed less tumors. CONCLUSION The immune response induced by H. nana infection is associated with the reduction of tumors probably due to the activity of eosinophils and neutrophils.
Collapse
Affiliation(s)
- E Ramos-Martínez
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - J Rojas-Serrano
- Servicio Clínico de enfermedades del Intersticio del Pulmón y Reumatología Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosío Villegas", Ciudad de México, Mexico
| | - O García-Hernández
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - F J García-Vázquez
- Instituto Nacional de Pediatría, Laboratorio de Inmunogenética Molecular, Departamento de Análisis Clínicos y Estudios Especiales, México, DF, Mexico
| | - W A Andrade
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - G Avila
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México. Ciudad de México, Mexico
| | - L Salinas-Pasquier
- Servicio de Anatomía Patológica, Unidad de Citopatología. Hospital General de México "Dr. Eduardo Liceaga", Ciudad de México, Mexico
| | - M R López-Vancell
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
43
|
Chambuso R, Ramesar R, Kaambo E, Denny L, Passmore JA, Williamson AL, Gray CM. Human Leukocyte Antigen (HLA) Class II -DRB1 and -DQB1 Alleles and the Association with Cervical Cancer in HIV/HPV Co-Infected Women in South Africa. J Cancer 2019; 10:2145-2152. [PMID: 31258717 PMCID: PMC6584421 DOI: 10.7150/jca.25600] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Background: A subset of women who are co-infected with Human Immunodeficiency Virus type 1 (HIV) and Human papillomavirus (HPV), progress rapidly to invasive cervical cancer regardless of antiretroviral therapy (ART) or immune status. We posit that HIV/HPV co-infection along with specific host HLA II -DRB1 and -DQB1 alleles play a major role in cervical cancer development. Methodology: We conducted a hospital-based genetic susceptibility case-control study in Cape Town, South Africa. We recruited 256 women of the same race, from which a total of 624 HLA-DRB1 and -DQB1 class II genotypes were studied. We characterized HLA II candidate genes using PCR based, Luminex intermediate resolution genotyping and confirmed significant associated genotypes at four-digit resolution by high resolution gel typing. We analyzed 160 alleles from cancer, 64 alleles from pre-cancer and 400 alleles from healthy control women. Whole blood was used for HIV antibody test and HLA II typing. Cervical tumor tissue biopsies were used for HPV genotyping. Tests were statistically significant if p<0.05. Results: Women who were co-infected with HIV/HPV had advanced cervical disease compared to women who were HIV negative. HLA class II -DQB1*03:01 and -DQB1*06:02 alleles were associated with cervical cancer in HIV/HPV co-infected women (p=0.001 and p<0.0001, respectively) while HLA class II -DRB1*13:01 and -DQB1*03:19 were rare or absent in women with cervical disease when compared to the control population (p=0.012 and 0.011, respectively). Conclusion: We describe associations between HLA class II genotypes with cervical cancer, or likely protection from cervical cancer disease in HIV/HPV co-infected South African women. Identifying mechanisms that give rise to this likely protective HLA association will provide insight into development of immune-based prevention measures.
Collapse
Affiliation(s)
- Ramadhani Chambuso
- MRC Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,Department of Gynaecology, Morogoro Regional Referral Hospital, Morogoro, Tanzania
| | - Raj Ramesar
- MRC Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Evelyn Kaambo
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,Department of Biochemistry and Medical Microbiology, University of Namibia School of Medicine, Windhoek, Namibia
| | - Lynette Denny
- Department of Obstetrics and Gynaecology, Groote Schuur Hospital, University of Cape Town, South Africa.,MRC/UCT Clinical Gynaecological Cancer Research Centre, Groote Schuur Hospital/University of Cape Town, South Africa
| | - Jo-Ann Passmore
- Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,MRC/UCT Clinical Gynaecological Cancer Research Centre, Groote Schuur Hospital/University of Cape Town, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,MRC/UCT Clinical Gynaecological Cancer Research Centre, Groote Schuur Hospital/University of Cape Town, South Africa
| | - Clive M Gray
- Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa.,Division of Immunology, Laboratory for Tissue Immunology, Department of Pathology and National Health Laboratory Service, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
44
|
Wang HF, Wang SS, Tang YJ, Chen Y, Zheng M, Tang YL, Liang XH. The Double-Edged Sword-How Human Papillomaviruses Interact With Immunity in Head and Neck Cancer. Front Immunol 2019; 10:653. [PMID: 31001266 PMCID: PMC6454067 DOI: 10.3389/fimmu.2019.00653] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/11/2019] [Indexed: 02/05/2023] Open
Abstract
Patients with human papilloma virus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) have remarkably better prognosis, which differs from HPV-negative oropharyngeal squamous cell carcinoma (OPSCC) with respect to clinical, genomic, molecular, and immunological aspects, especially having the characteristics of high levels of immune cell infiltration and high degrees of immunosuppression. This review will summarize immune evasion mechanisms in HPV-positive HNSCC, analyze the host various immune responses to HPV and abundant numbers of infiltrating immune cell, and discuss the differences between HPV-positive HNSCC with cervical cancer. A deeper understanding of the immune landscape will help new concepts to emerge in immune-checkpoint oncology, which might be a valuable add-on to established concepts.
Collapse
Affiliation(s)
- Hao-Fan Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Sha-Sha Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, Department of Oral Pathology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, Department of Oral Pathology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| |
Collapse
|
45
|
Paaso A, Jaakola A, Syrjänen S, Louvanto K. From HPV Infection to Lesion Progression: The Role of HLA Alleles and Host Immunity. Acta Cytol 2019; 63:148-158. [PMID: 30783048 DOI: 10.1159/000494985] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 10/29/2018] [Indexed: 01/27/2023]
Abstract
Persistent high-risk human papillomavirus (HPV) infection has been associated with increased risk for cervical precancerous lesions and cancer. The host's genetic variability is known to play a role in the development of cervical cancer. The human leukocyte antigen (HLA) genes are highly polymorphic and have shown to be important risk determinants of HPV infection persistence and disease progression. HLA class I and II cell surface molecules regulate the host's immune system by presenting HPV-derived peptides to T-cells. The activation of T-cell response may vary depending on the HLA allele polymorphism. The engagement of the T-cell receptor with the HPV peptide-HLA complex to create an active costimulatory signal is essential for the activation of the T-cell response. Functional peptide presentation by both HLA class I and II molecules is needed to activate efficient helper and effector T-cell responses in HPV infection recognition and clearance. Some of these HLA risk alleles could also be used as preventive tools in the detection of HPV-induced cervical lesions and cancer. These HLA alleles, together with HPV vaccines, could potentially offer possible solutions for reducing HPV-induced cervical cancer as well as other HPV-related cancers.
Collapse
Affiliation(s)
- Anna Paaso
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland,
- Department of Obstetrics and Gynecology, Turku University Hospital, University of Turku, Turku, Finland,
| | - Anna Jaakola
- Department of Obstetrics and Gynecology, Turku University Hospital, University of Turku, Turku, Finland
- Department of Obstetrics and Gynecology, Kymenlaakso Central Hospital, Kotka, Finland
| | - Stina Syrjänen
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pathology, Turku University Hospital, University of Turku, Turku, Finland
| | - Karolina Louvanto
- Department of Oral Pathology, Institute of Dentistry, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Obstetrics and Gynecology, Turku University Hospital, University of Turku, Turku, Finland
| |
Collapse
|
46
|
Efficacy of PD-1 blockade in cervical cancer is related to a CD8 +FoxP3 +CD25 + T-cell subset with operational effector functions despite high immune checkpoint levels. J Immunother Cancer 2019; 7:43. [PMID: 30755279 PMCID: PMC6373123 DOI: 10.1186/s40425-019-0526-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/31/2019] [Indexed: 01/08/2023] Open
Abstract
Background Cervical cancer (CxCa) is mainly a locally invading disease that metastasizes to loco-regional lymph node basins before involving distant organs in more advanced stages. Local immune potentiation of tumor-draining lymph nodes (TDLN) may thus protect against tumor progression. Methods To identify therapeutic targets for local immune modulation, multi-parameter flow cytometric T-cell profiling of primary cervical tumors (PT) and TDLN (n = 37) was performed. The in-vitro effect of PD-1 blockade on T-cell reactivity to HPV16 E6 oncoproteins was determined in cultures of TDLN and PT single cell suspensions (n = 19). Also, intracellular cytokine staining (ICS) upon anti-CD3 stimulation was performed in metastatic TDLN (LN+) and PT (n = 7), as well as multiplexed immunofluorescence histochemistry staining (n = 8). Results Our data revealed elevated rates of activated regulatory T cells (aTregs) and of central or effector memory CD8+ T cells in metastatic TDLN (LN+) as compared to tumor-free TDLN (LN-), and equally high or even higher rates of these subsets in PT. Both memory subsets co-expressed multiple immune checkpoints. PD-1 blockade significantly enhanced detectable E6-specific T-cell responses in 4/5 HPV16+ LN+ and in 1/5 HPV16+ PT. Whereas aTreg rates were higher in anti-PD-1 non-responders, in responders elevated levels of CD8+FoxP3+CD25+ T cells were observed, which correlated with the efficacy of PD-1 blockade (P = 0.018). This subset was characterized by an early effector memory phenotype with particularly high levels of co-expressed PD-1, CTLA-4, TIM-3 and LAG-3 checkpoints, but, rather than exhausted, was shown upon polyclonal activation to produce higher levels of Granzyme-B and effector cytokines as compared to its CD8+FoxP3− counterparts. Conclusion These observations support local PD-(L)1 blockade to interrupt loco-regional immune suppression in CxCa and control metastatic spread to TDLN. Furthermore, our data identify CD8+FoxP3+CD25+ T cells as therapeutic targets, which may also serve as predictive biomarker for PD-(L)1 checkpoint blockade. Electronic supplementary material The online version of this article (10.1186/s40425-019-0526-z) contains supplementary material, which is available to authorized users.
Collapse
|
47
|
Campbell-Tofte J, Vrahatis A, Josefsen K, Mehlsen J, Winther K. Investigating the aetiology of adverse events following HPV vaccination with systems vaccinology. Cell Mol Life Sci 2019; 76:67-87. [PMID: 30324425 PMCID: PMC11105185 DOI: 10.1007/s00018-018-2925-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 12/18/2022]
Abstract
In contrast to the insidious and poorly immunogenic human papillomavirus (HPV) infections, vaccination with the HPV virus-like particles (vlps) is non-infectious and stimulates a strong neutralizing-antibody response that protects HPV-naïve vaccinees from viral infection and associated cancers. However, controversy about alleged adverse events following immunization (AEFI) with the vlps have led to extensive reductions in vaccine acceptance, with countries like Japan dropping it altogether. The AEFIs are grouped into chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). In this review, we present a hypothesis that the AEFIs might arise from malfunctions within the immune system when confronted with the unusual antigen. In addition, we outline how the pathophysiology of the AEFIs can be cost-effectively investigated with the holistic principles of systems vaccinology in a two-step process. First, comprehensive immunological profiles of HPV vaccinees exhibiting the AEFIs are generated by integrating the data derived from serological profiling for prominent HPV antibodies and serum cytokines, with data from serum metabolomics, peripheral white blood cells transcriptomics and gut microbiome profiling. Next, the immunological profiles are compared with corresponding profiles generated for matched (a) HPV vaccinees without AEFIs; (b) non-HPV-vaccinated individuals with CFS/ME-like symptoms; and (c) non-HPV-vaccinated individuals without CFS/ME. In these comparisons, any causal links between HPV vaccine and the AEFIs, as well as the underlying molecular basis for the links will be revealed. Such a study should provide an objective basis for evaluating HPV vaccine safety and for identifying biomarkers for individuals at risk of developing AEFI with HPV vaccination.
Collapse
Affiliation(s)
| | | | - Knud Josefsen
- Bartholin Institute, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
| | - Jesper Mehlsen
- Coordinating Research Centre, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000, Frederiksberg, Denmark
| | - Kaj Winther
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Nørre Allé 51, DK-2200, Copenhagen N, Denmark
| |
Collapse
|
48
|
Jacome AA, Eng C. Experimental and investigational drugs for the treatment of anal cancer. Expert Opin Investig Drugs 2018; 27:941-950. [PMID: 30381968 DOI: 10.1080/13543784.2018.1543659] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Squamous cell carcinoma of the anal canal (SCCA) is a rare malignancy, but its incidence rates have been increasing in the last decade. Studies have demonstrated that up to 47% of patients with locally advanced disease have high-risk features for treatment failure. The potential high rates of recurrence after standard chemoradiotherapy for locally advanced disease and the lack of established care for metastatic disease have created an urgent need for the evaluation of new drugs that will ultimately improve the efficacy of treatment. AREAS COVERED This review presents results of recent phase-I and -II clinical trials which evaluate novel therapeutic modalities. The review also describes the findings of comprehensive genomic profiling studies which provide insights for promising therapeutics. EXPERT OPINION HPV vaccination is underutilized in the United States and as a result, HPV-associated malignancies are likely to continue for several decades; however, pivotal breakthroughs may create a foundation for distinctive treatment approaches for other HPV-associated malignancies for which no other standard of care exists.
Collapse
Affiliation(s)
- Alexandre A Jacome
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Cathy Eng
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
49
|
Abdulrahman Z, Kortekaas KE, De Vos Van Steenwijk PJ, Van Der Burg SH, Van Poelgeest MIE. The immune microenvironment in vulvar (pre)cancer: review of literature and implications for immunotherapy. Expert Opin Biol Ther 2018; 18:1223-1233. [DOI: 10.1080/14712598.2018.1542426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ziena Abdulrahman
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kim E Kortekaas
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sjoerd H Van Der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
50
|
Risk factors for cytological progression in HPV 16 infected women with ASC-US or LSIL: The Korean HPV cohort. Obstet Gynecol Sci 2018; 61:662-668. [PMID: 30474012 PMCID: PMC6236089 DOI: 10.5468/ogs.2018.61.6.662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/12/2018] [Accepted: 03/08/2018] [Indexed: 11/08/2022] Open
Abstract
Objective This study was to identify the risk factors for cytological progression in women with atypical squamous cells of undetermined significance (ASC-US) or low-grade squamous intraepithelial lesions (LSIL). Methods We analyzed data from women infected with the human papillomavirus (HPV) who participated in the Korean HPV cohort study. The cohort recruited women aged 20-60 years with abnormal cervical cytology (ASC-US or LSIL) from April 2010. All women were followed-up at every 6-month intervals with cervical cytology and HPV DNA testing. Results Of the 1,158 women included, 654 (56.5%) and 504 (43.5%) women showed ASC-US and LSIL, respectively. At the time of enrollment, 143 women tested positive for HPV 16 (85 single and 58 multiple infections). Cervical cytology performed in the HPV 16-positive women showed progression in 27%, no change in 23%, and regression in 50% of the women at the six-month follow-up. The progression rate associated with HPV 16 infection was higher than that with infection caused by other HPV types (relative risk [RR], 1.75; 95% confidence interval [CI], 1.08-2.84; P=0.028). The cytological progression rate in women with persistent HPV 16 infection was higher than that in women with incidental or cleared infections (P<0.001). Logistic regression analysis showed a significant relationship between cigarette smoking and cytological progression (RR, 4.15; 95% CI, 1.01-17.00). Conclusion The cytological progression rate in HPV 16-positive women with ASC-US or LSIL is higher than that in women infected with other HPV types. Additionally, cigarette smoking may play a role in cytological progression.
Collapse
|