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
|
Scurtu LG, Scurtu F, Dumitrescu SC, Simionescu O. Squamous Cell Carcinoma In Situ-The Importance of Early Diagnosis in Bowen Disease, Vulvar Intraepithelial Neoplasia, Penile Intraepithelial Neoplasia, and Erythroplasia of Queyrat. Diagnostics (Basel) 2024; 14:1799. [PMID: 39202286 PMCID: PMC11353497 DOI: 10.3390/diagnostics14161799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second-most-prevalent malignancy in humans. A delayed diagnosis of cSCC leads to heightened invasiveness and positive surgical margins. Bowen's disease (BD) represents an early form of cSCC and presents as a small erythematous, photo-distributed, psoriasiform plaque. Although certain dermoscopy features in BD are quite characteristic, histopathology remains the gold standard for diagnosis and provides a severity-scoring system that assists in guiding appropriate treatment strategies. The classification of precancerous lesions of the vulva and penis has undergone multifarious transformations due to variations in clinical and histopathological characteristics. Presently, erythroplasia of Queyrat is categorized as a clinical variant of penile intraepithelial neoplasia (PeIN). The diagnoses of vulvar intraepithelial neoplasia (VIN) and PeIN present significant challenges and typically necessitate one or more biopsies, potentially guided by dermoscopy. Aceto-white testing demonstrates a notably high negative predictive value for genital precancerous lesions. Histopathological examination represents the gold-standard diagnosis in VIN and PeIN, while p16 and p53 immunostainings alongside HPV testing provide crucial diagnostic clues. The histopathologic features, degree of differentiation, and associations with lichen planus, lichen sclerosus, and HPV guide the selection of conservative treatments or surgical excision.
Collapse
Affiliation(s)
- Lucian G. Scurtu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (L.G.S.); (S.C.D.)
- Department of Dermatology I, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Francesca Scurtu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (L.G.S.); (S.C.D.)
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011132 Bucharest, Romania
| | - Sebastian Catalin Dumitrescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (L.G.S.); (S.C.D.)
- Department of Dermatology I, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Olga Simionescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (L.G.S.); (S.C.D.)
- Department of Dermatology I, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
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
|
The common HLA class I-restricted tumor-infiltrating T cell response in HPV16-induced cancer. Cancer Immunol Immunother 2022; 72:1553-1565. [DOI: 10.1007/s00262-022-03350-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
AbstractImmunotherapies targeting truly tumor-specific targets focus on the expansion and activation of T cells against neoantigens or oncogenic viruses. One target is the human papilloma virus type 16 (HPV16), responsible for several anogenital cancers and oropharyngeal carcinomas. Spontaneous and vaccine-induced HPV-specific T cells have been associated with better clinical outcome. However, the epitopes and restriction elements to which these T cells respond remained elusive. To identify CD8+ T cell epitopes in cultures of tumor infiltrating lymphocytes, we here used multimers and/or a functional screening platform exploiting single HLA class I allele-engineered antigen presenting cells. This resulted in the detection of 20 CD8+ T cell responses to 11 different endogenously processed HLA-peptide combinations within 12 HPV16-induced tumors. Specific HLA-peptide combinations dominated the response in patients expressing these HLA alleles. T cell receptors (TCRs) reactive to seven different HLA class I-restricted peptides could be isolated and analysis revealed tumor reactivity for five of the six TCRs analyzed. The tumor reactive TCRs to these dominant HLA class I peptide combinations can potentially be used to engineer tumor-specific T cells for adoptive cell transfer approaches to treat HPV16-induced cancers.
Collapse
|
5
|
Gardella B, Gritti A, Soleymaninejadian E, Pasquali MF, Riemma G, La Verde M, Schettino MT, Fortunato N, Torella M, Dominoni M. New Perspectives in Therapeutic Vaccines for HPV: A Critical Review. Medicina (B Aires) 2022; 58:medicina58070860. [PMID: 35888579 PMCID: PMC9315585 DOI: 10.3390/medicina58070860] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/19/2022] [Accepted: 06/24/2022] [Indexed: 12/21/2022] Open
Abstract
Human Papillomavirus is the main cause of cervical cancer, including squamous cell carcinoma of the oropharynx, anus, rectum, penis, vagina, and vulva. In recent years, considerable effort has been made to control HPV-induced diseases using either prophylactic or therapeutic approaches. A critical review of the literature about the therapeutic Human Papillomavirus vaccine was performed to analyze its efficacy in the treatment of female lower genital tract lesions and its possible perspective application in clinical practice. The most important medical databases were consulted, and all papers published from 2000 until 2021 were considered. We retrieved a group of seven papers, reporting the role of anti HPV therapeutic vaccines against the L2 protein in the order of their efficacy and safety in female lower genital tract disease. In addition, the immune response due to vaccine administration was evaluated. The development of therapeutic vaccines represents an interesting challenge for the treatment of HPV infection of the lower genital tract. Literature data underline that the L2 protein may be an interesting and promising target in the development of therapeutic HPV vaccines, but the possible strengths and the unclear longevity of L2 immune responses are factors to be considered before clinical use.
Collapse
Affiliation(s)
- Barbara Gardella
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, 27100 Pavia, Italy; (B.G.); (M.F.P.); (M.D.)
- Department of Obstetrics and Gynecology, IRCCS Fundation Policlinico San Matteo, 27100 Pavia, Italy;
| | - Andrea Gritti
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, 27100 Pavia, Italy; (B.G.); (M.F.P.); (M.D.)
- Department of Obstetrics and Gynecology, IRCCS Fundation Policlinico San Matteo, 27100 Pavia, Italy;
- Correspondence: ; Tel.: +39-00382-503722
| | - Ehsan Soleymaninejadian
- Department of Obstetrics and Gynecology, IRCCS Fundation Policlinico San Matteo, 27100 Pavia, Italy;
| | - Marianna Francesca Pasquali
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, 27100 Pavia, Italy; (B.G.); (M.F.P.); (M.D.)
- Department of Obstetrics and Gynecology, IRCCS Fundation Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gaetano Riemma
- Obstetrics and Gynecology Unit, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy; (G.R.); (M.L.V.); (M.T.S.); (N.F.); (M.T.)
| | - Marco La Verde
- Obstetrics and Gynecology Unit, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy; (G.R.); (M.L.V.); (M.T.S.); (N.F.); (M.T.)
| | - Maria Teresa Schettino
- Obstetrics and Gynecology Unit, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy; (G.R.); (M.L.V.); (M.T.S.); (N.F.); (M.T.)
| | - Nicola Fortunato
- Obstetrics and Gynecology Unit, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy; (G.R.); (M.L.V.); (M.T.S.); (N.F.); (M.T.)
| | - Marco Torella
- Obstetrics and Gynecology Unit, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy; (G.R.); (M.L.V.); (M.T.S.); (N.F.); (M.T.)
| | - Mattia Dominoni
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, 27100 Pavia, Italy; (B.G.); (M.F.P.); (M.D.)
- Department of Obstetrics and Gynecology, IRCCS Fundation Policlinico San Matteo, 27100 Pavia, Italy;
| |
Collapse
|
6
|
Trutnovsky G, Reich O, Joura EA, Holter M, Ciresa-König A, Widschwendter A, Schauer C, Bogner G, Jan Z, Boandl A, Kalteis MS, Regauer S, Tamussino K. Topical imiquimod versus surgery for vulvar intraepithelial neoplasia: a multicentre, randomised, phase 3, non-inferiority trial. Lancet 2022; 399:1790-1798. [PMID: 35483400 DOI: 10.1016/s0140-6736(22)00469-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The optimal management of vulvar high-grade squamous intraepithelial lesions (vHSILs) is challenging. Surgery is the standard treatment, but recurrences are observed in half of patients. Medical treatment with imiquimod is an effective alternative, but the two modalities have not been compared in a randomised trial. The aim of this study was to compare the clinical effectiveness, histological response, human papillomavirus (HPV) clearance, acceptance, and psychosexual morbidity of primary imiquimod treatment versus surgical treatment in women with vHSIL. METHODS This study was a multicentre, randomised, phase 3, non-inferiority clinical trial done by the Austrian Gynaecological Oncology group at six hospitals in Austria. We recruited female patients aged 18-90 years with histologically confirmed vHSIL with visible unifocal or multifocal lesions. Main exclusion criteria were clinical suspicion of invasion, a history of vulvar cancer or severe inflammatory dermatosis of the vulva, and any active treatment for vHSIL within the previous 3 months. Women with known immunodeficiency, who were pregnant, or who were lactating were excluded. Patients were randomly assigned (1:1) by block randomisation to imiquimod or surgery, and stratified by unifocal or multifocal disease. Treatment with imiquimod was self-administered in a slowly escalating dosage scheme up to three times per week for a period of 4-6 months. Surgery consisted of excision or ablation. Patients were assessed with vulvoscopy, vulvar biopsy, HPV tests, and patient-reported outcomes at baseline and after 6 months and 12 months. The primary endpoint was complete clinical response (CCR) at 6 months after local imiquimod treatment or one surgical intervention. Primary analysis was per protocol with a non-inferiority margin of 20%. This trial is registered at ClinicalTrials.gov, NCT01861535. FINDINGS 110 patients with vHSIL (78% with unifocal vHSIL and 22% with multifocal vHSIL) were randomly assigned between June 7, 2013, and Jan 8, 2020. Clinical response to treatment could be assessed in 107 patients (54 in the imiquimod group and 53 in the surgery group), and 98 patients (46 in the imiquimod group and 52 in the surgery group) completed the study per protocol. 37 (80%) of 46 patients using imiquimod had CCR, compared with 41 (79%) of 52 patients after one surgical intervention, showing non-inferiority of the new treatment (difference in proportion -0·016, 95% CI -0·15 to -0·18; p=0·0056). Invasive disease was found in five patients at primary or secondary surgery, but not in patients with per-protocol imiquimod treatment. There was no significant difference in HPV clearance, adverse events, and treatment satisfaction between study groups. INTERPRETATION Imiquimod is a safe, effective, and well accepted alternative to surgery for women with vHSIL and can be considered as first-line treatment. FUNDING Austrian Science Fund and Austrian Gynaecological Oncology group.
Collapse
Affiliation(s)
- Gerda Trutnovsky
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria.
| | - Olaf Reich
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Elmar A Joura
- Department of Gynaecology and Gynaecological Oncology, Medical University of Vienna, Vienna, Austria
| | - Magdalena Holter
- Institute of Medical Informatics, Statistics, and Documentation, Medical University of Graz, Graz, Austria
| | - Alexandra Ciresa-König
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Widschwendter
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Schauer
- Department of Gynaecology, Krankenhaus Barmherzige Brüder Graz, Graz, Austria
| | - Gerhard Bogner
- Department of Obstetrics and Gynaecology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ziga Jan
- Department of Obstetrics and Gynaecology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Angelika Boandl
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Martin S Kalteis
- Department of Applied Tumour Biology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sigrid Regauer
- Department of Pathology, Medical University of Graz, Graz, Austria
| | - Karl Tamussino
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| |
Collapse
|
7
|
Rafael TS, Rotman J, Brouwer OR, van der Poel HG, Mom CH, Kenter GG, de Gruijl TD, Jordanova ES. Immunotherapeutic Approaches for the Treatment of HPV-Associated (Pre-)Cancer of the Cervix, Vulva and Penis. J Clin Med 2022; 11:1101. [PMID: 35207374 PMCID: PMC8876514 DOI: 10.3390/jcm11041101] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023] Open
Abstract
Human papillomavirus (HPV) infection drives tumorigenesis in almost all cervical cancers and a fraction of vulvar and penile cancers. Due to increasing incidence and low vaccination rates, many will still have to face HPV-related morbidity and mortality in the upcoming years. Current treatment options (i.e., surgery and/or chemoradiation) for urogenital (pre-)malignancies can have profound psychosocial and psychosexual effects on patients. Moreover, in the setting of advanced disease, responses to current therapies remain poor and nondurable, highlighting the unmet need for novel therapies that prevent recurrent disease and improve clinical outcome. Immunotherapy can be a useful addition to the current therapeutic strategies in various settings of disease, offering relatively fewer adverse effects and potential improvement in survival. This review discusses immune evasion mechanisms accompanying HPV infection and HPV-related tumorigenesis and summarizes current immunotherapeutic approaches for the treatment of HPV-related (pre-)malignant lesions of the uterine cervix, vulva, and penis.
Collapse
Affiliation(s)
- Tynisha S. Rafael
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (T.S.R.); (O.R.B.); (H.G.v.d.P.)
| | - Jossie Rotman
- Department of Obstetrics and Gynecology, Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.R.); (C.H.M.); (G.G.K.)
| | - Oscar R. Brouwer
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (T.S.R.); (O.R.B.); (H.G.v.d.P.)
| | - Henk G. van der Poel
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (T.S.R.); (O.R.B.); (H.G.v.d.P.)
| | - Constantijne H. Mom
- Department of Obstetrics and Gynecology, Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.R.); (C.H.M.); (G.G.K.)
| | - Gemma G. Kenter
- Department of Obstetrics and Gynecology, Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.R.); (C.H.M.); (G.G.K.)
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Ekaterina S. Jordanova
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (T.S.R.); (O.R.B.); (H.G.v.d.P.)
- Department of Obstetrics and Gynecology, Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.R.); (C.H.M.); (G.G.K.)
| |
Collapse
|
8
|
Bakker NAM, Rotman J, van Beurden M, Zijlmans HJM, van Ruiten M, Samuels S, Nuijen B, Beijnen J, De Visser K, Haanen J, Schumacher T, de Gruijl TD, Jordanova ES, Kenter GG, van den Berg JH, van Trommel NE. HPV-16 E6/E7 DNA tattoo vaccination using genetically optimized vaccines elicit clinical and immunological responses in patients with usual vulvar intraepithelial neoplasia (uVIN): a phase I/II clinical trial. J Immunother Cancer 2021; 9:jitc-2021-002547. [PMID: 34341131 PMCID: PMC8330588 DOI: 10.1136/jitc-2021-002547] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 11/23/2022] Open
Abstract
Background Usual vulvar intraepithelial neoplasia (uVIN) is a premalignancy caused by persistent infection with high-risk types of human papillomavirus (HPV), mainly type 16. Even though different treatment modalities are available (eg, surgical excision, laser evaporation or topical application of imiquimod), these treatments can be mutilating, patients often have recurrences and 2%–8% of patients develop vulvar carcinoma. Therefore, immunotherapeutic strategies targeting the pivotal oncogenic HPV proteins E6 and E7 are being explored to repress carcinogenesis. Method In this phase I/II clinical trial, 14 patients with HPV16+ uVIN were treated with a genetically enhanced DNA vaccine targeting E6 and E7. Safety, clinical responses and immunogenicity were assessed. Patients received four intradermal HPV-16 E6/E7 DNA tattoo vaccinations, with a 2-week interval, alternating between both upper legs. Biopsies of the uVIN lesions were taken at screening and +3 months after last vaccination. Digital photography of the vulva was performed at every check-up until 12 months of follow-up for measurement of the lesions. HPV16-specific T-cell responses were measured in blood over time in ex vivo reactivity assays. Results Vaccinations were well tolerated, although one grade 3 suspected unexpected serious adverse reaction was observed. Clinical responses were observed in 6/14 (43%) patients, with 2 complete responses and 4 partial responses (PR). 5/14 patients showed HPV-specific T-cell responses in blood, measured in ex vivo reactivity assays. Notably, all five patients with HPV-specific T-cell responses had a clinical response. Conclusions Our results indicate that HPV-16 E6/E7 DNA tattoo vaccination is a biologically active and safe treatment strategy in patients with uVIN, and suggest that T-cell reactivity against the HPV oncogenes is associated with clinical benefit. Trial registration number NTR4607.
Collapse
Affiliation(s)
- Noor Alida Maria Bakker
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Division of Tumor Biology and Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Jossie Rotman
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marc van Beurden
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Henry J Maa Zijlmans
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Maartje van Ruiten
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Sanne Samuels
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jos Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Karin De Visser
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - John Haanen
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Ton Schumacher
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, -Cancer Center Amsterdam, Amsterdam UMC-Vrije Universiteit, Amsterdam, The Netherlands
| | - Ekaterina S Jordanova
- Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gemma G Kenter
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joost H van den Berg
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Nienke E van Trommel
- Center for Gynecologic Oncology Amsterdam (CGOA), The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Abdulrahman Z, de Miranda NFCC, Hellebrekers BWJ, de Vos van Steenwijk PJ, van Esch EMG, van der Burg SH, van Poelgeest MIE. A pre-existing coordinated inflammatory microenvironment is associated with complete response of vulvar high-grade squamous intraepithelial lesions to different forms of immunotherapy. Int J Cancer 2020; 147:2914-2923. [PMID: 32574376 PMCID: PMC7540004 DOI: 10.1002/ijc.33168] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/12/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy of vulvar high-grade squamous intraepithelial lesion (vHSIL) is investigated as an alternative for surgery, because of high comorbidity and risk of recurrence. Limited evidence exists on the role and composition of the immune microenvironment in current immunotherapeutic approaches for vHSIL. The vHSIL of 29 patients biopsied before treatment with imiquimod were analyzed by two multiplex seven-color immunofluorescence panels to investigate the pre-existing T-cell and myeloid cell composition in relation to treatment response. The samples were scanned with the Vectra multispectral imaging system. Cells were automatically phenotyped and counted with inForm advanced image analysis software. Cell counts and composition were compared to that of vHSIL patients before therapeutic vaccination (n = 29) and to healthy vulva (n = 27). Our data show that the immune microenvironment of complete responders (CR) to imiquimod resembled the coordinated infiltration with type 1 CD4+ and CD8+ T cells and CD14+ inflammatory myeloid cells also found in healthy vulva. However, more CD8+ T cells and FoxP3+ regulatory T cells were present in CR. The lesions of partial responders (PR) lacked such a coordinated response and displayed an impaired influx of CD14+ inflammatory myeloid cells. Importantly, complete responses after imiquimod or therapeutic vaccination showed the same dependency on a pre-existing coordinated type 1 T-cell and CD14+ myeloid cell infiltration. In conclusion, a good clinical outcome after two different forms of immunotherapy for vHSIL is associated with the presence of a primary inflammatory process resulting in the coordinated influx of several types of immune cells which is then amplified.
Collapse
Affiliation(s)
- Ziena Abdulrahman
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Edith M G van Esch
- Department of Gynaecology, Catharina Hospital, Eindhoven, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
10
|
Lebreton M, Carton I, Brousse S, Lavoué V, Body G, Levêque J, Nyangoh-Timoh K. Vulvar intraepithelial neoplasia: Classification, epidemiology, diagnosis, and management. J Gynecol Obstet Hum Reprod 2020; 49:101801. [PMID: 32417455 DOI: 10.1016/j.jogoh.2020.101801] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 11/26/2022]
Abstract
Vulvar intraepithelial neoplasia (VIN) is classified into two entities: differentiated (dVIN) and vulvar high-grade squamous intraepithelial lesions (vH-SIL). dVIN is a premalignant lesion that develops on an existing vulvar lesion such as lichen sclerosus, while vH-SIL is associated with HPV infection. The two entities differ in terms of pathophysiology, background, prognosis, and management. The incidence of VIN in young women is rising and recurrence is common, even after radical surgery, which can cause significant disfigurement. Alternative strategies include topical treatments, ablation, and a watch-and-wait approach. There is currently no consensus on how these lesions should be managed. We review the literature in this field.
Collapse
Affiliation(s)
- M Lebreton
- Département de Gynécologie Obstétrique et Reproduction Humaine, CHU Anne de Bretagne, 16 Bd de Bulgarie BP 90347, F-35 203, Rennes Cedex 2, France
| | - I Carton
- Département de Gynécologie Obstétrique et Reproduction Humaine, CHU Anne de Bretagne, 16 Bd de Bulgarie BP 90347, F-35 203, Rennes Cedex 2, France
| | - S Brousse
- Département de Gynécologie Obstétrique et Reproduction Humaine, CHU Anne de Bretagne, 16 Bd de Bulgarie BP 90347, F-35 203, Rennes Cedex 2, France
| | - V Lavoué
- Département de Gynécologie Obstétrique et Reproduction Humaine, CHU Anne de Bretagne, 16 Bd de Bulgarie BP 90347, F-35 203, Rennes Cedex 2, France
| | - G Body
- Service de gynécologie obstétrique et médecine fœtale, université François Rabelais, CHRU de Tours, 2, boulevard Tonnelle, 37044, Tours Cedex 9, France
| | - J Levêque
- Département de Gynécologie Obstétrique et Reproduction Humaine, CHU Anne de Bretagne, 16 Bd de Bulgarie BP 90347, F-35 203, Rennes Cedex 2, France.
| | - K Nyangoh-Timoh
- Département de Gynécologie Obstétrique et Reproduction Humaine, CHU Anne de Bretagne, 16 Bd de Bulgarie BP 90347, F-35 203, Rennes Cedex 2, France
| |
Collapse
|
11
|
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: 22.8] [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
|
12
|
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: 2.2] [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
|
13
|
Cho JH, Lee HJ, Ko HJ, Yoon BI, Choe J, Kim KC, Hahn TW, Han JA, Choi SS, Jung YM, Lee KH, Lee YS, Jung YJ. The TLR7 agonist imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget 2018; 8:24932-24948. [PMID: 28212561 PMCID: PMC5421900 DOI: 10.18632/oncotarget.15326] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 01/23/2017] [Indexed: 12/31/2022] Open
Abstract
Toll-like receptor (TLR) ligands are strongly considered immune-adjuvants for cancer immunotherapy and have been shown to exert direct anti-cancer effects. This study was performed to evaluate the synergistic anti-cancer and anti-metastatic effects of the TLR7 agonist imiquimod (IMQ) during radiotherapy for melanoma. The pretreatment of B16F10 or B16F1 cells with IMQ combined with γ-ionizing radiation (IR) led to enhanced cell death via autophagy, as demonstrated by increased expression levels of autophagy-related genes, and an increased number of autophagosomes in both cell lines. The results also confirmed that the autophagy process was accelerated via the reactive oxygen species (ROS)-mediated MAPK and NF-κB signaling pathway in the cells pretreated with IMQ combined with IR. Mice subcutaneously injected with melanoma cells showed a reduced tumor growth rate after treatment with IMQ and IR. Treatment with 3-methyladenine (3-MA), ameliorated the anti-cancer effect of IMQ combined with IR. Additionally, the combination therapy enhanced anti-cancer immunity, as demonstrated by an increased number of CD8+ T cells and decreased numbers of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs) in the tumor lesions. Moreover, the combination therapy decreased the number of metastatic nodules in the lungs of mice that were injected with B16F10 cells via the tail vein. In addition, the combination therapy enhanced systemic anti-cancer immunity by increasing the abundances of T cell populations expressing IFN-γ and TNF-α. Therefore, these findings suggest that IMQ could serve as a radiosensitizer and immune booster during radiotherapy for melanoma patients.
Collapse
Affiliation(s)
- Jeong Hyun Cho
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyo-Ji Lee
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Byung-Il Yoon
- Department of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jongseon Choe
- Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Keun-Cheol Kim
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Tae-Wook Hahn
- Department of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jeong A Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Sun Shim Choi
- Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon, Republic of Korea
| | - Kee-Ho Lee
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, Republic of Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seodaemun-gu, Seoul, Republic of Korea
| | - Yu-Jin Jung
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
14
|
Medrano RF, Hunger A, Mendonça SA, Barbuto JAM, Strauss BE. Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy. Oncotarget 2017; 8:71249-71284. [PMID: 29050360 PMCID: PMC5642635 DOI: 10.18632/oncotarget.19531] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
During the last decades, the pleiotropic antitumor functions exerted by type I interferons (IFNs) have become universally acknowledged, especially their role in mediating interactions between the tumor and the immune system. Indeed, type I IFNs are now appreciated as a critical component of dendritic cell (DC) driven T cell responses to cancer. Here we focus on IFN-α and IFN-β, and their antitumor effects, impact on immune responses and their use as therapeutic agents. IFN-α/β share many properties, including activation of the JAK-STAT signaling pathway and induction of a variety of cellular phenotypes. For example, type I IFNs drive not only the high maturation status of DCs, but also have a direct impact in cytotoxic T lymphocytes, NK cell activation, induction of tumor cell death and inhibition of angiogenesis. A variety of stimuli, including some standard cancer treatments, promote the expression of endogenous IFN-α/β, which then participates as a fundamental component of immunogenic cell death. Systemic treatment with recombinant protein has been used for the treatment of melanoma. The induction of endogenous IFN-α/β has been tested, including stimulation through pattern recognition receptors. Gene therapies involving IFN-α/β have also been described. Thus, harnessing type I IFNs as an effective tool for cancer therapy continues to be studied.
Collapse
Affiliation(s)
- Ruan F.V. Medrano
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aline Hunger
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Samir Andrade Mendonça
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - José Alexandre M. Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Cell and Molecular Therapy Center, NUCEL-NETCEM, University of São Paulo, São Paulo, Brazil
| | - Bryan E. Strauss
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
15
|
Miles B, Safran HP, Monk BJ. Therapeutic options for treatment of human papillomavirus-associated cancers - novel immunologic vaccines: ADXS11-001. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2017; 4:10. [PMID: 28725449 PMCID: PMC5512733 DOI: 10.1186/s40661-017-0047-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/03/2017] [Indexed: 01/22/2023]
Abstract
Survival of patients with advanced, recurrent, or metastatic human papillomavirus (HPV)-associated cancer is suboptimal despite the availability of various treatment modalities. The recently developed bacterial vector Listeria monocytogenes (Lm) activates innate and adaptive immune responses and is expected to offer immunologic advantages. Axalimogene filolisbac (AXAL or ADXS11-001) is a novel immunotherapeutic based on the live, irreversibly attenuated Lm fused to the nonhemolytic fragment of listeriolysin O (Lm-LLO) and secretes the Lm-LLO-HPV E7 fusion protein targeting HPV-positive tumors. Herein are reported the development and recent results of various clinical trials in patients with HPV-associated cervical, head and neck, and anal cancers.
Collapse
Affiliation(s)
- Brett Miles
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Bradley J. Monk
- Division of Gynecologic Oncology, Arizona Oncology (US Oncology Network), University of Arizona College of Medicine, Creighton University School of Medicine at St. Joseph’s Hospital, 2222 E. Highland Ave, Suite 400, Phoenix, AZ 85016 USA
| |
Collapse
|
16
|
Torelli T, Catanzaro MA, Nicolai N, Giannatempo P, Necchi A, Raggi D, Paolini B, Colecchia M, Piva L, Biasoni D, Stagni S, Tesone A, Salvioni R. Treatment of Carcinoma In Situ of the Glans Penis With Topical Imiquimod Followed by Carbon Dioxide Laser Excision. Clin Genitourin Cancer 2017; 15:e483-e487. [DOI: 10.1016/j.clgc.2017.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/04/2017] [Accepted: 01/10/2017] [Indexed: 10/20/2022]
|
17
|
Woodby B, Scott M, Bodily J. The Interaction Between Human Papillomaviruses and the Stromal Microenvironment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:169-238. [PMID: 27865458 PMCID: PMC5727914 DOI: 10.1016/bs.pmbts.2016.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human papillomaviruses (HPVs) are small, double-stranded DNA viruses that replicate in stratified squamous epithelia and cause a variety of malignancies. Current efforts in HPV biology are focused on understanding the virus-host interactions that enable HPV to persist for years or decades in the tissue. The importance of interactions between tumor cells and the stromal microenvironment has become increasingly apparent in recent years, but how stromal interactions impact the normal, benign life cycle of HPVs, or progression of lesions to cancer is less understood. Furthermore, how productively replicating HPV impacts cells in the stromal environment is also unclear. Here we bring together some of the relevant literature on keratinocyte-stromal interactions and their impacts on HPV biology, focusing on stromal fibroblasts, immune cells, and endothelial cells. We discuss how HPV oncogenes in infected cells manipulate other cells in their environment, and, conversely, how neighboring cells may impact the efficiency or course of HPV infection.
Collapse
Affiliation(s)
- B Woodby
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - M Scott
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - J Bodily
- Louisiana State University Health Sciences Center, Shreveport, LA, United States.
| |
Collapse
|
18
|
van Poelgeest MIE, Welters MJP, Vermeij R, Stynenbosch LFM, Loof NM, Berends-van der Meer DMA, Löwik MJG, Hamming ILE, van Esch EMG, Hellebrekers BWJ, van Beurden M, Schreuder HW, Kagie MJ, Trimbos JBMZ, Fathers LM, Daemen T, Hollema H, Valentijn ARPM, Oostendorp J, Oude Elberink JHNG, Fleuren GJ, Bosse T, Kenter GG, Stijnen T, Nijman HW, Melief CJM, van der Burg SH. Vaccination against Oncoproteins of HPV16 for Noninvasive Vulvar/Vaginal Lesions: Lesion Clearance Is Related to the Strength of the T-Cell Response. Clin Cancer Res 2016; 22:2342-50. [PMID: 26813357 DOI: 10.1158/1078-0432.ccr-15-2594] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/03/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Therapeutic vaccination with human papillomavirus type 16 (HPV16) E6 and E7 synthetic long peptides (SLP) is effective against HPV16-induced high-grade vulvar and vaginal intraepithelial neoplasia (VIN/VaIN). However, clinical nonresponders displayed weak CD8(+) T-cell reactivity. Here, we studied if imiquimod applied at the vaccine site could improve CD8(+) T-cell reactivity, clinical efficacy, and safety of HPV16-SLP (ISA101). EXPERIMENTAL DESIGN A multicenter open-label, randomized controlled trial was conducted in patients with HPV16(+) high-grade VIN/VaIN. Patients received ISA101 vaccination with or without application of 5% imiquimod at the vaccine site. The primary objective was the induction of a directly ex vivo detectable HPV16-specific CD8(+) T-cell response. The secondary objectives were clinical responses (lesion size, histology, and virology) and their relation with the strength of vaccination-induced immune responses. RESULTS Forty-three patients were assigned to either ISA101 with imiquimod (n = 21) or ISA101 only (n = 22). Imiquimod did not improve the outcomes of vaccination. However, vaccine-induced clinical responses were observed in 18 of 34 (53%; 95% CI, 35.1-70.2) patients at 3 months and in 15 of 29 (52%; 95% CI, 32.5-70.6) patients, 8 of whom displayed a complete histologic response, at 12 months after the last vaccination. All patients displayed vaccine-induced T-cell responses, which were significantly stronger in patients with complete responses. Importantly, viral clearance occurred in all but one of the patients with complete histologic clearance. CONCLUSIONS This new study confirms that clinical efficacy of ISA101 vaccination is related to the strength of vaccine-induced HPV16-specific T-cell immunity and is an effective therapy for HPV16-induced high-grade VIN/VaIN. Clin Cancer Res; 22(10); 2342-50. ©2016 AACRSee related commentary by Karaki et al., p. 2317.
Collapse
Affiliation(s)
| | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Renee Vermeij
- Department of Gynaecology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Linda F M Stynenbosch
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Nikki M Loof
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Margriet J G Löwik
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ineke L E Hamming
- Department of Gynaecology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Edith M G van Esch
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Bart W J Hellebrekers
- Department of Obstetrics and Gynaecology, Haga Teaching Hospital, the Hague, the Netherlands
| | | | - Henk W Schreuder
- Department of Obstetrics and Gynaecology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Marjolein J Kagie
- Department of Obstetrics and Gynaecology, Medical Centre Haaglanden, the Hague, the Netherlands
| | - J Baptist M Z Trimbos
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Lorraine M Fathers
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Toos Daemen
- Department of Medical Microbiology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Harry Hollema
- Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, the Netherlands
| | - A Rob P M Valentijn
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jaap Oostendorp
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - J Hanneke N G Oude Elberink
- Department of Allergy, Internal Medicine, Groningen Research Institute for Asthma and COPD, University Medical Centre Groningen, Groningen, the Netherlands
| | - Gertjan J Fleuren
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gemma G Kenter
- Department of Gynaecology, Academic Medical Centre, Amsterdam, the Netherlands
| | - Theo Stijnen
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Hans W Nijman
- Department of Gynaecology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Cornelis J M Melief
- ISA Pharmaceuticals, Leiden, the Netherlands. Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
19
|
Scott ME, Ma Y, Farhat S, Moscicki AB. Expression of nucleic acid-sensing Toll-like receptors predicts HPV16 clearance associated with an E6-directed cell-mediated response. Int J Cancer 2015; 136:2402-8. [PMID: 25346143 PMCID: PMC4355215 DOI: 10.1002/ijc.29283] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/25/2014] [Indexed: 12/15/2022]
Abstract
Toll-like receptors (TLRs), important in rapid clearance of incident human papillomavirus (HPV) infections, may also be important in shaping the adaptive response to persistent infections. We examined here the association between TLR expression and clearance of HPV16 infections following periods of persistence, using longitudinal TLR measurements and a time-to-clearance analysis, as well as the interaction between TLRs and adaptive, cell-mediated responses involved in clearance. TLR2, TLR3, TLR7, TLR8 and TLR9 mRNA expression were measured in cervical cytobrush samples by quantitative PCR. Responses to the HPV16 E6 and E7 oncoproteins were measured by an interferon-γ immunospot assay. Bivariable and multivariable Cox proportional hazard models were used to estimate the effect of TLRs on HPV16 clearance. Higher expression of TLR3 or TLR7 at an HPV16-positive visit was a significant (p ≤ 0.05) predictor of clearance by the following visit, in both unadjusted and adjusted (for smoking and oral contraceptive use) models. In women with, but not those without, a positive response to E6, higher expression of TLR3 (hazard ratio: 1.2 [95% confidence interval: 1.04-1.39], p = 0.012), TLR7 (1.39 [1.14-1.7], p = 0.001), TLR8 (1.37 [1.11-1.69], p = 0.003), or TLR9 (1.53 [1.13-2.08], p = 0.006) was significantly associated with clearance, revealing an important link between innate and adaptive immunity in the control of HPV infections following periods of persistence.
Collapse
Affiliation(s)
- Mark E. Scott
- Division of Adolescent Medicine, Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Yifei Ma
- Division of Adolescent Medicine, Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Sepideh Farhat
- Division of Adolescent Medicine, Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Anna-Barbara Moscicki
- Division of Adolescent Medicine, Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, California, 94143, USA
| |
Collapse
|
20
|
Preti M, Igidbashian S, Costa S, Cristoforoni P, Mariani L, Origoni M, Sandri MT, Boveri S, Spolti N, Spinaci L, Sanvito F, Preti EP, Falasca A, Radici G, Micheletti L. VIN usual type-from the past to the future. Ecancermedicalscience 2015; 9:531. [PMID: 25987900 PMCID: PMC4431399 DOI: 10.3332/ecancer.2015.531] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Indexed: 12/27/2022] Open
Abstract
Usual vulvar intraepithelial neoplasia (uVIN) is the most common VIN type, generally related to a human papillomavirus (HPV) infection, predominantly type 16. The incidence of uVIN has been increasing over the last decades, and a bimodal peak is observed at the age of 40–44 and over 55 years. Almost 40% of patients with uVIN have a past, concomitant or future HPV-associated lesion of the lower genital tract. HPV-related malignancies are associated with a persistent HPV infection. The host immune response is of crucial importance in determining clearance or persistence of both HPV infections and HPV-related VIN. About 60% of the patients present with symptoms. Clinical features of uVIN vary in site, number, size, shape, colour, and thickness of lesions. Multicentric disease is often present. Most uVIN lesions are positive at immunohistochemistry to p16ink4a and p14arf, but negative to p53. Irrespective of surgical treatment used, uVIN recurrence rates are high. Positive margins do not predict the development of invasive disease and the need to re-excide the tissue around the scare remains to be demonstrated. Therefore, considering the low progression rate of uVIN and psycosexual sequelae, treatments should be as conservative as possible. Medical treatments available are mainly based on immunotherapy to induce normalisation of immune cell count in uVIN. None are approved by the food and drug administration (FDA) for the treatment of uVIN. If medical treatment is performed, adequate biopsies are required to reduce the risk of unrecognised invasive disease. Some studies suggest that failure to respond to immunotherapy might be related to a local immunosuppressive microenvironment, but knowledge of the uVIN microenvironment is limited. Moreover, our knowledge of the potential mechanisms involved in the escape of HPV-induced lesions from the immune system has many gaps. HPV vaccines have been demonstrated to be effective in preventing uVIN, with 94.9% efficacy in the HPV-naive population, while studies on therapeutic vaccines are limited. The low incidence of VIN requires large multicentre studies to determine the best way to manage affected patients and to investigate the immunological characteristics of the ‘vulvar microenviroment’ which leads to the persistence of HPV.
Collapse
Affiliation(s)
- Mario Preti
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy ; The Italian HPV Study Group (IHSG)
| | - Sarah Igidbashian
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Silvano Costa
- M.F. Toniolo Hospital, Bologna 40100, Italy ; The Italian HPV Study Group (IHSG)
| | - Paolo Cristoforoni
- Villa Montallegro, Genova 16100, Italy ; The Italian HPV Study Group (IHSG)
| | - Luciano Mariani
- HPV-Unit Gynecologic Oncology, Regina Elena National Cancer Institute of Rome, Rome 00100, Italy ; The Italian HPV Study Group (IHSG)
| | - Massimo Origoni
- Department of Obstetrics and Gynecology, Vita Salute San Raffaele University School of Medicine, Milano 20100, Italy ; The Italian HPV Study Group (IHSG)
| | - Maria T Sandri
- Division of Laboratory Medicine, European Institute of Oncology, Milano 20100, Italy ; The Italian HPV Study Group (IHSG)
| | - Sara Boveri
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Noemi Spolti
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Laura Spinaci
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Francesca Sanvito
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Eleonora P Preti
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Adriana Falasca
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Gianluigi Radici
- Preventive Gynecology Unit, European Institute of Oncology, Milano 20100, Italy
| | - Leonardo Micheletti
- Department of Obstetrics and Gynecology, University of Torino, Torino 10100, Italy
| |
Collapse
|
21
|
Antigen design enhances the immunogenicity of Semliki Forest virus-based therapeutic human papillomavirus vaccines. Gene Ther 2015; 22:560-7. [PMID: 25756550 DOI: 10.1038/gt.2015.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 01/21/2015] [Accepted: 02/23/2015] [Indexed: 01/09/2023]
Abstract
Cellular immunity against cancer can be achieved with viral vector- and DNA-based immunizations. In preclinical studies, cancer vaccines are very potent, but in clinical trials these potencies are not achieved yet. Thus, a rational approach to improve cancer vaccines is warranted. We previously demonstrated that the relatively low intrinsic immunogenicity of DNA vaccines could be enhanced by inclusion of endoplasmic reticulum (ER) targeting and universal helper epitopes within the vaccine. We now evaluated whether an optimal antigen format, as defined in DNA vaccines, can further enhance the effectiveness of recombinant Semliki Forest virus (rSFV) vaccines. To this purpose, we generated, characterized and evaluated the efficacy of rSFV replicon particles expressing human papillomavirus E6 and/or E7 proteins fused to several helper T-cell epitopes and an ER targeting signal. Here, we show that inclusion of a helper cassette and an ER targeting signal enhanced protein stability and markedly augmented the frequencies of human papillomavirus-specific T cells. Even at an immunization dose of as low as 10(5) replicon particles, this novel vaccine achieved tumor regression and protection. Thus, even highly effective viral vector vaccines can benefit from an improved antigen format, based on the inclusion of defined helper epitopes and ER targeting.
Collapse
|
22
|
Wang JW, Hung CF, Huh WK, Trimble CL, Roden RBS. Immunoprevention of human papillomavirus-associated malignancies. Cancer Prev Res (Phila) 2015; 8:95-104. [PMID: 25488410 PMCID: PMC4315720 DOI: 10.1158/1940-6207.capr-14-0311] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Persistent infection by one of 15 high-risk human papillomavirus (hrHPV) types is a necessary but not sufficient cause of 5% of all human cancers. This provides a remarkable opportunity for cancer prevention via immunization. Since Harald zur Hausen's pioneering identification of hrHPV types 16 and 18, found in approximately 50% and 20% of cervical cancers, respectively, two prophylactic HPV vaccines containing virus-like particles (VLP) of each genotype have been widely licensed. These vaccines are beginning to affect infection and HPV-associated neoplasia rates after immunization campaigns in adolescents. Here, we review recent progress and opportunities to better prevent HPV-associated cancers, including broadening immune protection to cover all hrHPV types, reducing the cost of HPV vaccines especially for developing countries that have the highest rates of cervical cancer, and immune-based treatment of established HPV infections. Screening based upon George Papanicolaou's cervical cytology testing, and more recently detection of hrHPV DNA/RNA, followed by ablative treatment of high-grade cervical intraepithelial neoplasia (CIN2/3) have substantially reduced cervical cancer rates, and we examine their interplay with immune-based modalities for the prevention and eventual elimination of cervical cancer and other HPV-related malignancies.
Collapse
Affiliation(s)
- Joshua W Wang
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland
| | - Chein-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland
| | - Warner K Huh
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Cornelia L Trimble
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland. Department of Oncology, The Johns Hopkins University, Baltimore, Maryland. Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, Maryland
| | - Richard B S Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland. Department of Oncology, The Johns Hopkins University, Baltimore, Maryland. Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
23
|
Peng S, Wang JW, Karanam B, Wang C, Huh WK, Alvarez RD, Pai SI, Hung CF, Wu TC, Roden RBS. Sequential cisplatin therapy and vaccination with HPV16 E6E7L2 fusion protein in saponin adjuvant GPI-0100 for the treatment of a model HPV16+ cancer. PLoS One 2015; 10:e116389. [PMID: 25560237 PMCID: PMC4283968 DOI: 10.1371/journal.pone.0116389] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Abstract
Clinical studies suggest that responses to HPV16 E6E7L2 fusion protein (TA-CIN) vaccination alone are modest, and GPI-0100 is a well-tolerated, potent adjuvant. Here we sought to optimize both the immunogenicity of TA-CIN via formulation with GPI-0100 and treatment of HPV16+ cancer by vaccination after cisplatin chemotherapy. HPV16 neutralizing serum antibody titers, CD4+ T cell proliferative and E6/E7-specific CD8+ T cell responses were significantly enhanced when mice were vaccinated subcutaneously (s.c.) or intramuscularly (i.m.) with TA-CIN formulated with GPI-0100. Vaccination was tested for therapy of mice bearing syngeneic HPV16 E6/E7+ tumors (TC-1) either in the lung or subcutaneously. Mice treated with TA-CIN/GPI-0100 vaccination exhibited robust E7-specific CD8+ T cell responses, which were associated with reduced tumor burden in the lung, whereas mice receiving either component alone were similar to controls. Since vaccination alone was not sufficient for cure, mice bearing s.c. TC-1 tumor were first treated with two doses of cisplatin and then vaccinated. Vaccination with TA-CIN/GPI-0100 i.m. substantially retarded tumor growth and extended survival after cisplatin therapy. Injection of TA-CIN alone, but not GPI-0100, into the tumor (i.t.) was similarly efficacious after cisplatin therapy, but the mice eventually succumbed. However, tumor regression and extended remission was observed in 80% of the mice treated with cisplatin and then intra-tumoral TA-CIN/GPI-0100 vaccination. These mice also exhibited robust E7-specific CD8+ T cell and HPV16 neutralizing antibody responses. Thus formulation of TA-CIN with GPI-0100 and intra-tumoral delivery after cisplatin treatment elicits potent therapeutic responses in a murine model of HPV16+ cancer.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Joshua W. Wang
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Balasubramanyam Karanam
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biology and Center for Cancer Research, Carver Research Foundation, Tuskegee University, Tuskegee, Alabama, United States of America
| | - Chenguang Wang
- Department of Biostatistics, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Warner K. Huh
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ronald D. Alvarez
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Sara I. Pai
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Chien-fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - T. -C. Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Richard B. S. Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
24
|
van Esch EMG, van Poelgeest MIE, Trimbos JBMZ, Fleuren GJ, Jordanova ES, van der Burg SH. Intraepithelial macrophage infiltration is related to a high number of regulatory T cells and promotes a progressive course of HPV-induced vulvar neoplasia. Int J Cancer 2014; 136:E85-94. [PMID: 25220265 DOI: 10.1002/ijc.29173] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/11/2014] [Accepted: 08/04/2014] [Indexed: 12/17/2022]
Abstract
Human papilloma virus (HPV)-induced usual-type vulvar intraepithelial neoplasia (uVIN) is infiltrated by myeloid cells but the type and role of these cells is unclear. We used triple immunofluorescent confocal microscopy to locate, identify and quantify myeloid cells based on their staining pattern for CD14, CD33 and CD163 in a cohort of 43 primary and 20 recurrent uVIN lesions, 21 carcinomas and 26 normal vulvar tissues. The progressive course of uVIN is characterized by an increase in both intraepithelial and stromal mature M1 and M2 macrophages. While the M2 macrophages outnumber M1 macrophages in healthy controls and uVIN, they are matched in number by M1 macrophages in cancer. Importantly, uVIN patients with a dense intraepithelial infiltration with mature CD14+ macrophages (irrespective of M1 or M2 type) displayed approximately a six times higher risk to develop a recurrence and a high number of these cells constituted an independent prognostic factor for recurrence. In addition, a dense intraepithelial CD14+ cell infiltration was associated with high numbers of intraepithelial CD4+ Tregs and low numbers of stromal CD8+TIM3+ T cells. Patients with low numbers of intraepithelial CD14+ cells and high numbers of stromal CD8+TIM3+ cells showed the best recurrence-free survival. These data clearly show the importance of the local immune response in HPV-induced vulvar neoplasia and may be of help in predicting the prognosis of patients or their response to immunotherapy.
Collapse
Affiliation(s)
- Edith M G van Esch
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
25
|
van Esch EMG, van Poelgeest MIE, Kouwenberg S, Osse EM, Trimbos JBMZ, Fleuren GJ, Jordanova ES, van der Burg SH. Expression of coinhibitory receptors on T cells in the microenvironment of usual vulvar intraepithelial neoplasia is related to proinflammatory effector T cells and an increased recurrence-free survival. Int J Cancer 2014; 136:E95-106. [PMID: 25220367 DOI: 10.1002/ijc.29174] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/11/2014] [Accepted: 08/04/2014] [Indexed: 02/04/2023]
Abstract
Human papillomavirus-induced usual-type vulvar intraepithelial neoplasia (uVIN) are infiltrated by immune cells but apparently not cleared. A potential explanation for this is an impaired T cell effector function by an immunesuppressive milieu, coinfiltrating regulatory T cells or the expression of coinhibitory molecules. Here, the role of these potential inhibitory mechanisms was evaluated by a detailed immunohistochemical analysis of T cell infiltration in the context of FoxP3, Tbet, indoleamine 2,3-dioxygenase, programmed cell death 1, T cell immunoglobulin mucin 3 (TIM3), natural killer cell lectin-like receptor A (NKG2A) and galectins-1, -3 and -9. Paraffin-embedded tissues of primary uVIN lesions (n=43), recurrent uVIN lesions (n=20), vulvar carcinoma (n=21) and healthy vulvar tissue (n=26) were studied. We show that the vulva constitutes an area intensely surveyed by CD8+, CD4+, Tbet+ and regulatory T cell populations, parts of which express the examined coinhibitory molecules. In uVIN especially, the number of regulatory T cells and TIM3+ T cells increased. The expression of the coinhibitory markers TIM3 and NKG2A probably reflected a higher degree of T cell activation as a dense infiltration with stromal CD8+TIM3+ T cells and CD3+NKG2A+ T cells was related to the absence of recurrences and/or a prolonged recurrence-free survival. A dense coinfiltrate with regulatory T cells was negatively associated with the time to recurrence, most dominantly when the stromal CD8+TIM3+ infiltration was limited. This notion was sustained in vulvar carcinoma's where the numbers of regulatory T cells progressively increased to outnumber coinfiltrating CD8+TIM3+ T cells and CD3+NKG2A+ T cells.
Collapse
Affiliation(s)
- Edith M G van Esch
- Department of Gynaecology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Human papillomavirus 16 E2-, E6- and E7-specific T-cell responses in children and their mothers who developed incident cervical intraepithelial neoplasia during a 14-year follow-up of the Finnish Family HPV cohort. J Transl Med 2014; 12:44. [PMID: 24524328 PMCID: PMC3929154 DOI: 10.1186/1479-5876-12-44] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/14/2014] [Indexed: 02/01/2023] Open
Abstract
Background Human papillomavirus (HPV) infection has traditionally been regarded as a sexually transmitted disease (STD), but recent evidence implicates that an infected mother can transmit HPV to her newborn during pregnancy, at delivery, perinatal period or later. Given the lack of any studies on HPV-specific immune responses in children, we conducted HPV16-specific cell-mediated immune (CMI) monitoring of the mother-child pairs with known oral and genital HPV follow-up (FU) data since the delivery. In the Finnish Family HPV Study, 10 out of 331 mothers developed incident cervical intraepithelial neoplasia (CIN) during their 14-year FU. Our hypothesis according to the common dogma is that there is no HPV16 specific immune response in offspring of the CIN mother as she/he has not started the sexual life yet. Methods We used overlapping 30–35 mer peptides covering the entire HPV16 E2, E6 and E7 protein sequences. Assays for lymphocyte proliferation capacity, cytokine production and HPV16-specific Foxp3 + CD25 + CD4+ regulatory T-cells were performed. Results HPV16-specific proliferative T-cell responses were broader in children than in their mothers. Nine of 10 children had responses against both E2 peptide pools compared to only 4 of the 10 mothers. Six of the 10 children and only 2 mothers displayed reactivity to E6 and/or E7. The cytokine levels of IL-2 (p = 0.023) and IL-5 (p = 0.028) induced by all peptide pools, were also higher among children than their mothers. The children of the mothers with incident CIN3 had significantly higher IFN-γ (p = 0.032) and TNF-α (p = 0.008) levels than other children. Conclusions Our study is the first to show that also children could have HPV-specific immunity. These data indicate that the children have circulating HPV16-specific memory T-cells which might have been induced by previous HPV16 exposure or ongoing HPV 16 infection.
Collapse
|
27
|
van Esch EMG, Tummers B, Baartmans V, Osse EM, Ter Haar N, Trietsch MD, Hellebrekers BWJ, Holleboom CAG, Nagel HTC, Tan LT, Fleuren GJ, van Poelgeest MIE, van der Burg SH, Jordanova ES. Alterations in classical and nonclassical HLA expression in recurrent and progressive HPV-induced usual vulvar intraepithelial neoplasia and implications for immunotherapy. Int J Cancer 2014; 135:830-42. [PMID: 24415578 DOI: 10.1002/ijc.28713] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/17/2013] [Indexed: 01/23/2023]
Abstract
Immunotherapy of usual vulvar intraepithelial neoplasia (uVIN) is promising; however, many patients still fail to show clinical responses, which could be explained by an immune escape through alterations in human leukocyte antigen (HLA) expression. Therefore, we analyzed a cohort of patients with a primary (n = 43) and subsequent recurrent uVIN lesion (n = 20), vaccine-treated uVIN patients (n = 12), patients with human papillomavirus (HPV)-induced vulvar carcinoma (n = 21) and healthy controls (n = 26) for the expression of classical HLA-class I/II and nonclassical HLA-E/-G and MHC class I chain-related molecule A (MICA). HLA-class I was downregulated in 70% of uVIN patients, including patients with a clinical response to immunotherapy. Downregulation of HLA-class I is probably reversible, as only 15% of the uVIN cases displayed loss of heterozygosity (LOH) and HLA-class I could be upregulated in uVIN keratinocyte cultures by interferon γ. HLA-class I downregulation is more frequently associated with LOH in vulvar carcinomas (25-55.5%). HLA-class II was found to be focally expressed in 65% of uVIN patients. Of the nonclassical molecules, MICA was downregulated in 80% of uVIN whereas HLA-E and -G were expressed in a minority of cases. Their expression was more prominent in vulvar carcinoma. No differences were found between the alterations observed in paired primary and recurrent uVIN. Importantly, downregulation of HLA-B/C in primary uVIN lesions was associated with the development of recurrences and progression to cancer. We conclude that downregulation of HLA is frequently observed in premalignant HPV-induced lesions, including clinical responders to immunotherapy, and is associated with worse clinical outcome. However, in the majority of cases downregulation may still be reversible.
Collapse
Affiliation(s)
- E M G van Esch
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands; Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Clinical Characteristics Associated With Development of Recurrence and Progression in Usual-Type Vulvar Intraepithelial Neoplasia. Int J Gynecol Cancer 2013; 23:1476-83. [DOI: 10.1097/igc.0b013e3182a57fd6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
29
|
Tong WWY, Hillman RJ, Kelleher AD, Grulich AE, Carr A. Anal intraepithelial neoplasia and squamous cell carcinoma in HIV-infected adults. HIV Med 2013; 15:65-76. [PMID: 24007498 DOI: 10.1111/hiv.12080] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2013] [Indexed: 12/25/2022]
Abstract
Anal cancer is one of the most common non-AIDS-defining malignancies in the era of combination antiretroviral therapy. Its precursor lesion, anal intraepithelial neoplasia (AIN), is highly prevalent in HIV-infected populations. More than 90% of anal squamous cell cancers are attributable to human papillomavirus (HPV). While the biology of HPV-related intraepithelial neoplasia is consistent across lower anogenital sites, the natural history of AIN is not well established and cannot be assumed to be identical to that of cervical intraepithelial neoplasia. Screening strategies to prevent anal cancer should be developed based on robust natural history data in HIV-infected and uninfected populations. Likewise, treatments need to be tested in randomized clinical trials, and reserved for those at significant risk of progression to cancer. This review covers the epidemiology, pathogenesis and immunology of HPV infection, AIN and anal cancer, and summarizes the current diagnosis, screening and treatment strategies in HIV-infected adults.
Collapse
Affiliation(s)
- W W Y Tong
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | | | | | | | | |
Collapse
|
30
|
Zhou Q, Zhu K, Cheng H. Toll-like receptors in human papillomavirus infection. Arch Immunol Ther Exp (Warsz) 2013; 61:203-15. [PMID: 23435874 DOI: 10.1007/s00005-013-0220-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 02/13/2013] [Indexed: 01/02/2023]
Abstract
Infection with human papillomaviruses (HPVs) often causes cutaneous benign lesions, cervical cancer, and a number of other tumors. The mechanisms of host immune system to prevent and control HPV infection still remain poorly understood. Toll-like receptors (TLRs) are specific pattern recognition molecules that bind to microbial components to trigger innate immunity and direct adaptive immunity in the face of immunological danger. TLRs have been established to play an essential role in sensing and initiating antiviral immune responses. Recent accumulating evidence demonstrated that HPVs modulate TLR expression and interfere with TLR signaling pathways, leading to persistent viral infection and carcinogenesis. This review summarizes current knowledge on the roles of TLR during HPV infection, focusing on TLR recognition, modulation of TLR expression and signaling, regulatory receptors involved in TLR signaling, and cross-talk of TLRs with antimicrobial peptides. Immunotherapeutic strategies based on TLR agonists have emerged to be one of the novel promising avenues in treatment of HPV-associated diseases in the future.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun Road East, Hangzhou 310016, People's Republic of China
| | | | | |
Collapse
|
31
|
van der Burg SH. Immunotherapy of human papilloma virus induced disease. Open Virol J 2012; 6:257-63. [PMID: 23341861 PMCID: PMC3547504 DOI: 10.2174/1874357901206010257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/28/2012] [Accepted: 08/17/2012] [Indexed: 01/08/2023] Open
Abstract
Immunotherapy is the generic name for treatment modalities aiming to reinforce the immune system against diseases in which the immune system plays a role. The design of an optimal immunotherapeutic treatment against chronic viruses and associated diseases requires a detailed understanding of the interactions between the target virus and its host, in order to define the specific strategies that may have the best chance to deliver success at each stage of disease. Recently, a first series of successes was reported for the immunotherapy of Human Papilloma Virus (HPV)-induced premalignant diseases but there is definitely room for improvement. Here I discuss a number of topics that in my opinion require more study as the answers to these questions allows us to better understand the underlying mechanisms of disease and as such to tailor treatment.
Collapse
Affiliation(s)
- Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Stern PL, van der Burg SH, Hampson IN, Broker TR, Fiander A, Lacey CJ, Kitchener HC, Einstein MH. Therapy of human papillomavirus-related disease. Vaccine 2012; 30 Suppl 5:F71-82. [PMID: 23199967 PMCID: PMC4155500 DOI: 10.1016/j.vaccine.2012.05.091] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/10/2012] [Accepted: 05/03/2012] [Indexed: 12/18/2022]
Abstract
This chapter reviews the current treatment of chronic and neoplastic human papillomavirus (HPV)-associated conditions and the development of novel therapeutic approaches. Surgical excision of HPV-associated lower genital tract neoplasia is very successful but largely depends on secondary prevention programmes for identification of disease. Only high-risk HPV-driven chronic, pre-neoplastic lesions and some very early cancers cannot be successfully treated by surgical procedures alone. Chemoradiation therapy of cervical cancer contributes to the 66-79% cervical cancer survival at 5 years. Outlook for those patients with persistent or recurrent cervical cancer following treatment is very poor. Topical agents such as imiquimod (immune response modifier), cidofovir (inhibition of viral replication; induction apoptosis) or photodynamic therapy (direct damage of tumour and augmentation of anti-tumour immunity) have all shown some useful efficacy (~50-60%) in treatment of high grade vulvar intraepithelial neoplasia (VIN). Provider administered treatments of genital warts include cryotherapy, trichloracetic acid, or surgical removal which has the highest primary clearance rate. Patient applied therapies include podophyllotoxin and imiquimod. Recurrence after "successful" treatment is 30-40%. Further improvements could derive from a rational combination of current therapy with new drugs targeting molecular pathways mediated by HPV in cancer. Small molecule inhibitors targeting the DNA binding activities of HPV E1/E2 or the anti-apoptotic consequences of E6/E7 oncogenes are in preclinical development. Proteasome and histone deacetylase inhibitors, which can enhance apoptosis in HPV positive tumour cells, are being tested in early clinical trials. Chronic high-risk HPV infection/neoplasia is characterised by systemic and/or local immune suppressive regulatory or escape factors. Recently two E6/E7 vaccines have shown some clinical efficacy in high grade VIN patients and this correlated with strong and broad systemic HPV-specific T cell response and modulation of key local immune factors. Treatments that can shift the balance of immune effectors locally in combination with vaccination are now being tested. This article forms part of a special supplement entitled "Comprehensive Control of HPV Infections and Related Diseases" Vaccine Volume 30, Supplement 5, 2012.
Collapse
Affiliation(s)
- Peter L Stern
- Paterson Institute for Cancer Research, University of Manchester, Manchester M20 4BX, UK.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
van Esch EMG, Welters MJP, Jordanova ES, Trimbos JBMZ, van der Burg SH, van Poelgeest MIE. Treatment failure in patients with HPV 16-induced vulvar intraepithelial neoplasia: understanding different clinical responses to immunotherapy. Expert Rev Vaccines 2012; 11:821-40. [PMID: 22913259 DOI: 10.1586/erv.12.56] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Failure of the immune system to launch a strong and effective immune response to high-risk HPV is related to viral persistence and the development of anogenital (pre)malignant lesions such as vulvar intraepithelial neoplasia (VIN). Different forms of immunotherapy, aimed at overcoming the inertia of the immune system, have been developed and met with clinical success. Unfortunately these, in principal successful, therapeutic approaches also fail to induce clinical responses in a substantial number of cases. In this review, the authors summarize the traits of the immune response to HPV in healthy individuals and in patients with HPV-induced neoplasia. The potential mechanisms involved in the escape of HPV-induced lesions from the immune system indicate gaps in our knowledge. Finally, the interaction between the immune system and VIN is discussed with a special focus on the different forms of immunotherapy applied to treat VIN and the potential causes of therapy failure. The authors conclude that there are a number of pre-existing conditions that determine the patients' responsiveness to immunotherapy. An immunotherapeutic strategy in which different aspects of immune failure are attacked by complementary approaches, will improve the clinical response rate.
Collapse
Affiliation(s)
- Edith M G van Esch
- Department of Gynecology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
34
|
Zom GGP, Khan S, Filippov DV, Ossendorp F. TLR ligand-peptide conjugate vaccines: toward clinical application. Adv Immunol 2012; 114:177-201. [PMID: 22449782 DOI: 10.1016/b978-0-12-396548-6.00007-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Approaches to treat cancer with therapeutic vaccination have made significant progress. In order to induce efficient antitumor immunity, a vaccine should target and activate antigen-presenting cells, such as the dendritic cell, while delivering the tumor-derived antigen of choice. Conjugates of synthetic peptides and ligands of pattern-recognition receptors (PRRs) combine these features and, given their synthetic nature, can be produced under GMP conditions. Therefore, conjugation of antigenic peptides to potent PRR ligands is a promising vaccination approach for the treatment of cancer. This review focuses on the different PRR families that can be exploited for the design of conjugates and explores the results obtained so far with PRR ligands conjugated to antigen. The uptake and processing of Toll-like receptor ligand-peptide conjugates are discussed in more detail, as well as future directions that may further enhance the immunogenicity of conjugates.
Collapse
Affiliation(s)
- Gijs G P Zom
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | |
Collapse
|
35
|
Jacobelli S, Sanaa F, Moyal-Barracco M, Pelisse M, Berville S, Villefroy P, North MO, Figueiredo S, Charmeteau B, Clerici T, Plantier F, Arnold F, Touzé A, Dupin N, Avril MF, Guillet JG, Cheynier R, Bourgault-Villada I. Anti-HPV16 E2 protein T-cell responses and viral control in women with usual vulvar intraepithelial neoplasia and their healthy partners. PLoS One 2012; 7:e36651. [PMID: 22590583 PMCID: PMC3348873 DOI: 10.1371/journal.pone.0036651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 04/11/2012] [Indexed: 11/19/2022] Open
Abstract
T-cell responses (proliferation, intracellular cytokine synthesis and IFNγ ELISPOT) against human papillomavirus 16 (HPV16) E2 peptides were tested during 18 months in a longitudinal study in eight women presenting with HPV16-related usual vulvar intraepithelial neoplasia (VIN) and their healthy male partners. In six women, anti-E2 proliferative responses and cytokine production (single IFNγ and/or dual IFNγ/IL2 and/or single IL2) by CD4+ T lymphocytes became detectable after treating and healing of the usual VIN. In the women presenting with persistent lesions despite therapy, no proliferation was observed. Anti-E2 proliferative responses were also observed with dual IFNγ/IL2 production by CD4+ T-cells in six male partners who did not exhibit any genital HPV-related diseases. Ex vivo IFNγ ELISPOT showed numerous effector T-cells producing IFNγ after stimulation by a dominant E2 peptide in all men and women. Since the E2 protein is absent from the viral particles but is required for viral DNA replication, these results suggest a recent infection with replicative HPV16 in male partners. The presence of polyfunctional anti-E2 T-cell responses in the blood of asymptomatic men unambiguously establishes HPV infection even without detectable lesions. These results, despite the small size of the studied group, provide an argument in favor of prophylactic HPV vaccination of young men in order to prevent HPV16 infection and viral transmission from men to women.
Collapse
Affiliation(s)
- Simon Jacobelli
- Institut Cochin, Université Paris Descartes, CNRS, UMR 8104, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Singh SK, Meyering M, Ramwadhdoebe TH, Stynenbosch LFM, Redeker A, Kuppen PJK, Melief CJM, Welters MJP, van der Burg SH. The simultaneous ex vivo detection of low-frequency antigen-specific CD4+ and CD8+ T-cell responses using overlapping peptide pools. Cancer Immunol Immunother 2012; 61:1953-63. [PMID: 22491788 PMCID: PMC3493661 DOI: 10.1007/s00262-012-1251-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 03/21/2012] [Indexed: 11/25/2022]
Abstract
The ability to measure antigen-specific T cells at the single-cell level by intracellular cytokine staining (ICS) is a promising immunomonitoring tool and is extensively applied in the evaluation of immunotherapy of cancer. The protocols used to detect antigen-specific CD8+ T-cell responses generally work for the detection of antigen-specific T cells in samples that have undergone at least one round of in vitro pre-stimulation. Application of a common protocol but now using long peptides as antigens was not suitable to simultaneously detect antigen-specific CD8+ and CD4+ T cells directly ex vivo in cryopreserved samples. CD8 T-cell reactivity to monocytes pulsed with long peptides as antigens ranged between 5 and 25 % of that observed against monocytes pulsed with a direct HLA class I fitting minimal CTL peptide epitope. Therefore, we adapted our ICS protocol and show that the use of tenfold higher concentration of long peptides to load APC, the use of IFN-α and poly(I:C) to promote antigen processing and improve T-cell stimulation, does allow for the ex vivo detection of low-frequency antigen-specific CD8+ and CD4+ T cells in an HLA-independent setting. While most of the improvements were related to increasing the ability to measure CD8+ T-cell reactivity following stimulation with long peptides to at least 50 % of the response detected when using a minimal peptide epitope, the final analysis of blood samples from vaccinated patients successfully showed that the adapted ICS protocol also increases the ability to ex vivo detect low-frequency p53-specific CD4+ T-cell responses in cryopreserved PBMC samples.
Collapse
Affiliation(s)
- Satwinder Kaur Singh
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO box 9600, 2300 RC Leiden, The Netherlands
| | - Maaike Meyering
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO box 9600, 2300 RC Leiden, The Netherlands
| | - Tamara H. Ramwadhdoebe
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO box 9600, 2300 RC Leiden, The Netherlands
| | - Linda F. M. Stynenbosch
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO box 9600, 2300 RC Leiden, The Netherlands
| | - Anke Redeker
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis J. M. Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J. P. Welters
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO box 9600, 2300 RC Leiden, The Netherlands
| | - Sjoerd H. van der Burg
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
37
|
Scholten KBJ, Turksma AW, Ruizendaal JJ, van den Hende M, van der Burg SH, Heemskerk MHM, Meijer CJLM, Hooijberg E. Generating HPV specific T helper cells for the treatment of HPV induced malignancies using TCR gene transfer. J Transl Med 2011; 9:147. [PMID: 21892941 PMCID: PMC3176193 DOI: 10.1186/1479-5876-9-147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 09/05/2011] [Indexed: 02/02/2023] Open
Abstract
Background Infection with high risk Human Papilloma Virus (HPV) is associated with cancer of the cervix, vagina, penis, vulva, anus and some cases of head and neck carcinomas. The HPV derived oncoproteins E6 and E7 are constitutively expressed in tumor cells and therefore potential targets for T cell mediated adoptive immunotherapy. Effective immunotherapy is dependent on the presence of both CD4+ and CD8+ T cells. However, low precursor frequencies of HPV16 specific T cells in patients and healthy donors hampers routine isolation of these cells for adoptive transfer purposes. An alternative to generate HPV specific CD4+ and CD8+ T cells is TCR gene transfer. Methods HPV specific CD4+ T cells were generated using either a MHC class I or MHC class II restricted TCR (from clones A9 and 24.101 respectively) directed against HPV16 antigens. Functional analysis was performed by interferon-γ secretion, proliferation and cytokine production assays. Results Introduction of HPV16 specific TCRs into blood derived CD4+ recipient T cells resulted in recognition of the relevant HPV16 epitope as determined by IFN-γ secretion. Importantly, we also show recognition of the endogenously processed and HLA-DP1 presented HPV16E6 epitope by 24.101 TCR transgenic CD4+ T cells and recognition of the HLA-A2 presented HPV16E7 epitope by A9 TCR transgenic CD4+ T cells. Conclusion Our data indicate that TCR transfer is feasible as an alternative strategy to generate human HPV16 specific CD4+ T helper cells for the treatment of patients suffering from cervical cancer and other HPV16 induced malignancies.
Collapse
Affiliation(s)
- Kirsten B J Scholten
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Therapeutic vaccination against human papilloma virus induced malignancies. Curr Opin Immunol 2011; 23:252-7. [DOI: 10.1016/j.coi.2010.12.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 12/23/2010] [Indexed: 11/23/2022]
|
39
|
Karim R, Meyers C, Backendorf C, Ludigs K, Offringa R, van Ommen GJB, Melief CJM, van der Burg SH, Boer JM. Human papillomavirus deregulates the response of a cellular network comprising of chemotactic and proinflammatory genes. PLoS One 2011; 6:e17848. [PMID: 21423754 PMCID: PMC3056770 DOI: 10.1371/journal.pone.0017848] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 02/10/2011] [Indexed: 02/06/2023] Open
Abstract
Despite the presence of intracellular pathogen recognition receptors that allow
infected cells to attract the immune system, undifferentiated keratinocytes
(KCs) are the main targets for latent infection with high-risk human papilloma
viruses (hrHPVs). HPV infections are transient but on average last for more than
one year suggesting that HPV has developed means to evade host immunity. To
understand how HPV persists, we studied the innate immune response of
undifferentiated human KCs harboring episomal copies of HPV16 and 18 by
genome-wide expression profiling. Our data showed that the expression of the
different virus-sensing receptors was not affected by the presence of HPV.
Poly(I:C) stimulation of the viral RNA receptors TLR3,
PKR, MDA5 and RIG-I, the
latter of which indirectly senses viral DNA through non-self RNA polymerase III
transcripts, showed dampening in downstream signalling of these receptors by
HPVs. Many of the genes downregulated in HPV-positive KCs involved components of
the antigen presenting pathway, the inflammasome, the production of antivirals,
pro-inflammatory and chemotactic cytokines, and components downstream of
activated pathogen receptors. Notably, gene and/or protein interaction analysis
revealed the downregulation of a network of genes that was strongly
interconnected by IL-1β, a crucial cytokine to activate adaptive immunity.
In summary, our comprehensive expression profiling approach revealed that HPV16
and 18 coordinate a broad deregulation of the keratinocyte's inflammatory
response, and contributes to the understanding of virus persistence.
Collapse
Affiliation(s)
- Rezaul Karim
- Center for Human and Clinical Genetics, Leiden
University Medical Center, Leiden, The Netherlands
- Department of Immunohematology and Blood
Transfusion, Leiden University Medical Center, Leiden, The
Netherlands
- Department of Clinical Oncology, Leiden
University Medical Center, Leiden, The Netherlands
| | - Craig Meyers
- Department of Microbiology and Immunology, The
Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United
States of America
| | - Claude Backendorf
- Laboratory of Molecular Genetics, Leiden
Institute of Chemistry, Gorlaeus Laboratories, Leiden University, Leiden, The
Netherlands
| | - Kristina Ludigs
- Department of Immunohematology and Blood
Transfusion, Leiden University Medical Center, Leiden, The
Netherlands
| | - Rienk Offringa
- Department of Immunohematology and Blood
Transfusion, Leiden University Medical Center, Leiden, The
Netherlands
| | - Gert-Jan B. van Ommen
- Center for Human and Clinical Genetics, Leiden
University Medical Center, Leiden, The Netherlands
| | - Cornelis J. M. Melief
- Department of Immunohematology and Blood
Transfusion, Leiden University Medical Center, Leiden, The
Netherlands
| | - Sjoerd H. van der Burg
- Department of Clinical Oncology, Leiden
University Medical Center, Leiden, The Netherlands
| | - Judith M. Boer
- Center for Human and Clinical Genetics, Leiden
University Medical Center, Leiden, The Netherlands
- Netherlands Bioinformatics Centre, Nijmegen,
The Netherlands
- * E-mail:
| |
Collapse
|
40
|
Gaisa MM, Goldstone SE. Diagnosis and Treatment of Anal Intraepithelial Neoplasia and Condylomata. SEMINARS IN COLON AND RECTAL SURGERY 2011. [DOI: 10.1053/j.scrs.2010.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
41
|
Daud II, Scott ME, Ma Y, Shiboski S, Farhat S, Moscicki AB. Association between toll-like receptor expression and human papillomavirus type 16 persistence. Int J Cancer 2011; 128:879-86. [PMID: 20473890 DOI: 10.1002/ijc.25400] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mechanisms involved in mucosal immune control of cervical human papillomavirus (HPV) infection remain ill defined. Because toll-like receptors (TLRs) are key players in innate immune responses, we investigated the association between TLR expression and viral persistence or clearance in young women with incident infections with oncogenic HPV types 16 or 51. Messenger RNA expression of TLR1, TLR2, TLR3, TLR4, TLR6, TLR7, TLR8 and TLR9 was measured by quantitative reverse transcription-PCR using human endocervical specimens, collected before and after viral acquisition, in a cohort well characterized for HPV infections. Wilcoxon rank sum test was used to compare the change seen from preinfection to incident infection between women who subsequently cleared infection with those who did not. HPV 16 infections that cleared were significantly (p < 0.05) associated with an increase in expression of the four viral nucleic acid-sensing TLRs (TLR3, TLR7, TLR8 and TLR9) as well as TLR2 upon viral acquisition. Similar associations were not observed for HPV 51. In women who subsequently cleared their HPV 16 infection, changes in TLR1, TLR3, TLR7 and TLR8 expression levels between preincident and incident visits were significantly correlated with parallel changes in the levels of interferon-α2, measured by immunoassay in cervical lavage specimens. This study suggests that dampened TLR expression in the cervical mucosa is a type-specific mechanism by which HPV 16 interferes with innate immune responses, contributing to viral persistence, and that TLR upregulation and resultant cytokine induction is important in subsequent viral clearance.
Collapse
Affiliation(s)
- Ibrahim I Daud
- Infectious Disease Research Training Program, University of California, San Francisco, CA 94118, USA
| | | | | | | | | | | |
Collapse
|
42
|
Terlou A, van Seters M, Ewing PC, Aaronson NK, Gundy CM, Heijmans-Antonissen C, Quint WGV, Blok LJ, van Beurden M, Helmerhorst TJM. Treatment of vulvar intraepithelial neoplasia with topical imiquimod: seven years median follow-up of a randomized clinical trial. Gynecol Oncol 2011; 121:157-62. [PMID: 21239049 DOI: 10.1016/j.ygyno.2010.12.340] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 12/15/2010] [Accepted: 12/16/2010] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Recently we reported on the efficacy of imiquimod for treating vulvar intraepithelial neoplasia (VIN) in a placebo-controlled, double-blinded randomized clinical trial (RCT). Four weeks after treatment, a complete response was observed in 35% of patients and a partial response in 46%. All complete responders remained disease-free at 12 months follow-up. In the current investigations, we assessed long-term follow-up at least 5 years after the initial RCT. METHODS Twenty-four of 26 imiquimod-treated patients who had participated in the initial RCT were seen for follow-up. Primary endpoint was durability of clinical response to imiquimod assessed by naked eye vulvar examination and histology. Long-term clinical response was correlated to lesion size before start of the initial RCT. Secondary endpoints were mental health, global quality of life, body image and sexual function in relation with long-term clinical response. RESULTS Median follow-up period was 7.2 years (range 5.6-8.3 years). VIN recurred in one of nine complete responders. Of the initial partial responders, two became disease-free after additional imiquimod treatment. In the other partial responders, VIN recurred at least once after the initial RCT. In long-term complete responders, lesion size at study entry was smaller and these patients had a significantly better global quality of life at follow-up than patients with residual disease and/or recurrence after imiquimod treatment. CONCLUSIONS In case of a complete response, imiquimod is effective in the long-term. Furthermore, patients with a long-term complete response had a significantly better global quality of life than patients who recurred after imiquimod treatment.
Collapse
Affiliation(s)
- Annelinde Terlou
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Centre, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
van der Burg SH, Arens R, Melief CJM. Immunotherapy for persistent viral infections and associated disease. Trends Immunol 2011; 32:97-103. [PMID: 21227751 DOI: 10.1016/j.it.2010.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 01/30/2023]
Abstract
Persistent viral infections reflect a failure of the host's immune system to control infection, and in many cases, they are associated with the development of malignancies. So far, vaccines designed to boost viral immunity during chronic infection have not been successful. Infections with high-risk human papilloma viruses (e.g. HPV16) are acquired by a large segment of the population and persist in 5-10% of infected individuals, which causes the development of high-grade pre-malignant lesions. Recently we succeeded in causing regression of HPV16-induced disease in ∼50% of chronically infected patients by a novel therapeutic vaccine. Here, we summarize the parallels in immunity against HPV and other chronic viruses and discuss the general implications of our findings for the immunotherapy of chronic infections.
Collapse
Affiliation(s)
- Sjoerd H van der Burg
- Department of Clinical Oncology, Building 1, K1-P, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands.
| | | | | |
Collapse
|
44
|
Daayana S, Winters U, Stern PL, Kitchener HC. Clinical and immunological response to photodynamic therapy in the treatment of vulval intraepithelial neoplasia. Photochem Photobiol Sci 2011; 10:802-9. [DOI: 10.1039/c0pp00344a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
45
|
Non-responders to topical imiquimod followed by vaccination therapy in VIN patients may be due to the level of IL10. Br J Cancer 2010; 103:595-6. [PMID: 20664597 PMCID: PMC2939795 DOI: 10.1038/sj.bjc.6605824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
46
|
Abstract
Anal squamous intraepithelial lesions include both low-grade squamous intraepithelial lesions (LSIL) and high-grade squamous intraepithelial lesions (HSIL) and are caused by chronic infection with the human papillomavirus (HPV). The disease is increasing in both incidence and prevalence, especially among patients with the following risk factors: homosexual men, acquired or iatrogenic immunosuppression, and presence of other HPV-related diseases. Although the natural history of the disease is unknown, there is significant evidence that untreated HSIL progresses to squamous cell carcinoma in 11% of patients and in up to 50% of patients with extensive disease and immunosuppression. Anal cytology and reflex HPV DNA testing are used to screen for disease, particularly among patients with the aforementioned risk factors. Evaluation of the patient should include physical examination and high-resolution anoscopy (HRA) to evaluate for disease above and below the dentate line. Intervention is warranted and this can be achieved in many ways. The treatment option associated with the best outcomes is ablation directed with HRA, which can be performed in the office or in the operating room with minimal morbidity. This strategy is effective in patients with both low-volume and high-volume disease and is associated with a malignant progression rate of 0.4% in patients with treated HSIL.
Collapse
Affiliation(s)
- Carlos E Pineda
- Section of Colon and Rectal Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA. cepineda@ stanford.edu
| | | |
Collapse
|
47
|
Abstract
Anal squamous intraepithelial lesions include both low-grade squamous intraepithelial lesions (LSIL) and high-grade squamous intraepithelial lesions (HSIL) and are caused by chronic infection with the human papillomavirus (HPV). The disease is increasing in both incidence and prevalence, especially among patients with the following risk factors: homosexual men, acquired or iatrogenic immunosuppression, and presence of other HPV-related diseases. Although the natural history of the disease is unknown, there is significant evidence that untreated HSIL progresses to squamous cell carcinoma in 11% of patients and in up to 50% of patients with extensive disease and immunosuppression. Anal cytology and reflex HPV DNA testing are used to screen for disease, particularly among patients with the aforementioned risk factors. Evaluation of the patient should include physical examination and high-resolution anoscopy (HRA) to evaluate for disease above and below the dentate line. Intervention is warranted and this can be achieved in many ways. The treatment option associated with the best outcomes is ablation directed with HRA, which can be performed in the office or in the operating room with minimal morbidity. This strategy is effective in patients with both low-volume and high-volume disease and is associated with a malignant progression rate of 0.4% in patients with treated HSIL.
Collapse
Affiliation(s)
- Carlos E Pineda
- Section of Colon and Rectal Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA. cepineda@ stanford.edu
| | | |
Collapse
|
48
|
Success or failure of vaccination for HPV16-positive vulvar lesions correlates with kinetics and phenotype of induced T-cell responses. Proc Natl Acad Sci U S A 2010; 107:11895-9. [PMID: 20547850 DOI: 10.1073/pnas.1006500107] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
One half of a group of 20 patients with human papillomavirus type 16 (HPV16)-induced vulvar intraepithelial neoplasia grade 3 displayed a complete regression (CR) after therapeutic vaccination with HPV16 E6/E7 synthetic long peptides. Patients with relatively larger lesions generally did not display a CR. To investigate immune correlates of treatment failure, patients were grouped according to median lesion size at study entry, and HPV16-specific immunity was analyzed at different time points by complementary immunological assays. The group of patients with smaller lesions displayed stronger and broader vaccine-prompted HPV16-specific proliferative responses with higher IFNgamma (P = 0.0003) and IL-5 (P < 0.0001) levels than patients with large lesions. Characteristically, this response was accompanied by a distinct peak in cytokine levels after the first vaccination. In contrast, the patient group with larger lesions mounted higher frequencies of HPV16-specific CD4(+)CD25(+)Foxp3(+) T cells (P = 0.005) and displayed a lower HPV16-specific IFNgamma/IL-10 ratio after vaccination (P < 0.01). No disparity in T memory immunity to control antigens was found, indicating that the differences in HPV-specific immunity did not reflect general immune failure. We observed a strong correlation between a defined set of vaccine-prompted specific immune responses and the clinical efficacy of therapeutic vaccination. Notably, a high ratio of HPV16-specific vaccine-prompted effector T cells to HPV16-specific CD4(+)CD25(+)Foxp3(+) T cells was predictive of clinical success. Foxp3(+) T cells have been associated previously with impaired immunity in malignancies. Here we demonstrate that the vaccine-prompted level of this population is associated with early treatment failure.
Collapse
|
49
|
Mahto M, Nathan M, O'Mahony C. More than a decade on: review of the use of imiquimod in lower anogenital intraepithelial neoplasia. Int J STD AIDS 2010; 21:8-16. [PMID: 20029061 DOI: 10.1258/ijsa.2009.009309] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To assess the effectiveness of 5% imiquimod cream (IQ) in the treatment of vulvar, penile and anal intraepithelial neoplasias (VIN, PIN and AIN), we searched Medline, Embase, PubMed and Cochrane Library databases. With regard to VIN there were two randomized controlled trials (RCTs), eight uncontrolled/cohort studies, nine case reports and one review article. Use of IQ in PIN and AIN were only supported by cohort studies (two each for PIN and AIN) and case reports (15 for PIN and 3 for AIN). On pooled analysis of RCTs, uncontrolled and cohort studies, the mean complete response (CR) rate for VIN, PIN and AIN were 51%, 70% and 48%, respectively. The mean partial response (PR) rate for VIN, PIN and AIN were 25%, 30% and 34% respectively. The recurrence (RR) rate for VIN, PIN and AIN were 16%, 0% and 36%, respectively. The follow-up period for VIN, PIN and AIN ranged from 2 to 32 months, 10 to 12 months and 11 to 39 months, respectively. Although the results for PIN look the best, the strongest evidence regarding efficacy of IQ in anogenital intraepithelial neoplasia is for VIN supported by RCTs. Evidence for use of IQ in AIN was essentially limited to HIV-positive men who have sex with men. IQ was reasonably well tolerated with side-effects being managed with reduction in frequency of drug usage and/or rest periods. Based on these results, IQ seems to be a safe mode of treatment and is possibly an alternative to currently available methods of treatment. However, there are no comparative studies assessing its efficacy against traditional modes of treatment.
Collapse
Affiliation(s)
- M Mahto
- Department of Genitourinary Medicine, Cheshire East Community Health (Central and Eastern Cheshire PCT), Assura Health and Wellness Centre, Sunderland Street, Macclesfield SK11 6JL.
| | | | | |
Collapse
|
50
|
Daayana S, Elkord E, Winters U, Pawlita M, Roden R, Stern PL, Kitchener HC. Phase II trial of imiquimod and HPV therapeutic vaccination in patients with vulval intraepithelial neoplasia. Br J Cancer 2010; 102:1129-36. [PMID: 20234368 PMCID: PMC2853099 DOI: 10.1038/sj.bjc.6605611] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Vulval intraepithelial neoplasia (VIN) is a premalignant condition, which is frequently associated with type HPV16 infection, and multifocal disease has high rates of surgical treatment failure. METHODS We report a phase II clinical trial of the topical immunomodulator, imiquimod, for 8 weeks, followed by 3 doses (weeks 10, 14 and 18) of therapeutic human papillomavirus (HPV) vaccination (TA-CIN, fusion protein HPV16 E6E7L2) in 19 women with VIN grades 2 and 3. Histology and HPV testing of biopsies were performed at weeks 0, 10, 20 and 52. Intralesional infiltration of T-cell subsets and lymphocyte proliferation for HPV systemic immune responses were also assessed. RESULTS Lesion response (complete regression of VIN on histology) was observed in 32% (6 out of 19) of women at week 10, increasing to 58% (11 out of 19) at week 20 and 63% (12 out of 19) at week 52. At this time, 36% (5 out of 14) of lesions showed HPV16 clearance and 79% (15 out of 19) of women were symptom free. At week 20, after treatment with imiquimod and vaccination, there was significantly increased local infiltration of CD8 and CD4 T cells in lesion responders; in contrast, non-responders (persistent VIN by histology) showed an increased density of T regulatory cells. After vaccination, only lesion responders had significantly increased lympho-proliferation to the HPV vaccine antigens. CONCLUSION The therapeutic effect of treatment depends on the differential immune response of responders and non-responders with affect locally and systemically.
Collapse
Affiliation(s)
- S Daayana
- Academic Unit of Obstetrics and Gynaecology, University of Manchester, St Mary's Hospital, Whitworth Park, Manchester M13 0JH, UK
| | | | | | | | | | | | | |
Collapse
|