Review Article
The Therapeutic Vaccine: Is it Feasible?

https://doi.org/10.1016/j.arcmed.2009.07.003Get rights and content

In contrast to the prophylactic HPV vaccines that exhibit great promise in reducing the burden of cervical cancer, there is limited progress towards the development of immune therapeutic strategies that would help those women who are already infected with high-risk HPV and do not benefit from the current vaccines. The reason for this drawback is the lack of knowledge about the immune mechanisms that control the growth of HPV-infected or -transformed cells in vivo. It became evident that the preclinical models in rodents provide only limited information about the performance of a candidate vaccine in humans. In particular, the immune correlate for a clinical response remains to be determined. On the other hand, HPV-related malignancies provide an excellent model for cancer immune therapies in general. There is hope that the continuous efforts of academic research combined with corporate involvement will finally present an efficient product.

Introduction

High-risk papillomaviruses are able to initiate persistent infections. From the perspective of a virus with low replication capacity (like HPV16), this is a reasonable strategy to ensure spread within a population and thus to survive. On the other hand, the ability to persist within a cell in the absence of virus replication that typically is associated with cell death is the prerequisite for the capacity of a given virus to transform a cell into uncontrolled growth and, as a consequence, induce cancer.

Papillomaviruses have developed different strategies to escape the immune control and thus establish a persistent infection. Typically the immune system does not recognize papillomaviruses because they remain restricted to the affected epithelium, i.e., there is no viremic spread to other organs. There is also low expression of viral proteins within the basal and first suprabasal layer in the absence of cell death, hence without induction of danger signals that would alert the innate immune system. Virus production occurs in the superficial, differentiated cells with limited or no interaction with the immune system.

In addition to this low-profile strategy, human papillomaviruses are also able to actively suppress the antiviral effect of the immune system, most effectively already at the level of the innate response. This suggestion stems from the results of a clinical study that demonstrated the inverse correlation between the expression of the E7 gene within a cervical lesion and its response to interferon (1). This observation was corroborated by experimental studies that demonstrated the interference of the E6 and E7 proteins of HPV 16 and 18 with the expression of TLR 9 (2) and type I interferons as well as with downstream events within the interferon-induced pathways (for review see Reference 3). Another immune-inhibitory effect of the viral oncoproteins is the inhibition of migration of APCs from the epithelium as required to exert their function within the lymph nodes 4, 5 and the strong reduction of the amount of surface MHC I molecules that are necessary for specific recognition by cytotoxic T cells (6).

Despite the immunosuppressive capacities and the hiding of the virus, the immune system nevertheless does participate in the control of HPV infections. There is an increased risk of HPV infections and associated lesions in immunosuppressed patients. For example, HIV-positive women with reduced CD4+ cell count are up to 8-fold more likely to develop a persistent genital HPV infection and a cervical dysplasia (7). Also, in some of the HPV-infected individuals, virus-specific T helper and/or cytotoxic T cells can be detected that are typically directed against the early proteins E2, E6 and E7 (for summary see References 8, 9, 10) and are often associated with the regression of the lesion. In regressing warts, infiltrating macrophages, NK cells and/or CD4+ lymphocytes have been detected 11, 12. Successful treatment with interferon of HPV-induced anal lesions goes along with infiltration by T-helper 1 cells that are characteristic for a delayed-type hypersensitivity reaction. When treatment fails, the patients showed a depletion of Langerhans' cells, reduction of MHC II expression in keratinocytes and of the cytokines IL-1a, -1b and GM-CSF (13). Similar observations have been made in patients after treatment with an immune modulator (imiquimod) that stimulates the innate immune system via TLR 7 14, 15. A humoral immune response against early HPV proteins can only occasionally be detected during the natural course of an infection and occurs typically only in patients with HPV-related malignant diseases 16, 17, 18. Measurable antibodies directed against the viral structural proteins occur in about half of the infected subjects and are considered a marker for virus persistence.

Section snippets

Experimental Models for HPV-specific Immune Therapy

Immune therapy of papillomavirus-induced lesions proved to be successful in case of BPV 4-induced warts in calves that were immunized with recombinant E7 protein after experimental infection (19). Regression of warts was also observed after immunization with the L2 minor structural protein. This result was unexpected because late proteins are thought to be confined to the superficial layers of differentiating epithelium distant to the influence of the immune system (20). Similarly surprising

Therapeutic Vaccines

The recently introduced prophylactic vaccines consisting of HPV16 or 18 virus-like particles (VLP) proved effective in preventing persistent infections and precancerous lesions (29). It is to be expected that they will substantially reduce the incidence of cervical cancer. Because it will take many years before this goal can be reached (30), it is important to develop strategies for immune therapy for those women who are already infected and thus have been shown not to benefit from the

Proteins

Purified proteins are inefficient in inducing CD8+ T-cell response as they are mostly processed through the MHC class II pathway, which directs the immune response towards the Th2 phenotype. Addition of appropriate adjuvants can prevent this bias 39, 40; however, only very few are licensed for use in humans.

Immunization with fusion proteins of HPV16 E7 linked to a variety of other proteins such as the Haemophilus influenzae lipoprotein D, the Mycobacterium bovis hsp65 or calreticulin 41, 42, 43

Peptides

Application of short peptides derived from tumor antigens is a straightforward strategy to induce a clinically relevant T-cell response (for review see Reference 50). The major advantage of this approach is based on the easy chemical synthesis thus high purity of the manufactured peptides.

The initial peptide-based immunotherapies in patients used HLA-A2 restricted CTL epitopes together with a universal HLA-DR helper epitope 51, 52, 53 (for review see Reference 54). Despite the induction of

Recombinant Vectors

Recombinant vaccinia (rVaccinia) is the prototype for vector-mediated delivery of HPV genes. Vaccinia itself induces a strong immune response and provides adjuvant effects 22, 59. HPV16 + 18 E6/E7 recombinant vaccinia were already tested in several clinical studies 22, 37, 41, 60, 61, 62, 63. A slightly modified vector based upon a vaccinia strain that has been used in human (modified virus Ankara: MVA) contains in addition to the viral gene the IL-2 gene that is expected to be of

Autologous DCs

Murine DCs loaded with protein were shown to induce strong T-cell responses upon inoculation into syngeneic animals 67, 68, 69. Similar effects were observed when DCs of HPV16 or 18 positive cervical cancer patients were loaded with the corresponding E7 protein leading to the in vitro induction of E7-specific Th1 CD8 + CTL and CD4+ responses against autologous tumor cells and cultured MHC I-matched cervical cancer cell 70, 71. In a study of 15 stage IV cervical cancer patients, inoculation of

Conclusions

Whereas the development of prophylactic vaccinations against certain human papilloma viruses aiming at the prevention of cervical cancer via neutralizing antibodies has been successfully completed, progress in immune therapies for treatment of HPV-related lesions is lagging behind. Of the ∼40 published studies, some yielded promising clinical responses in a portion of patients, yet there is no clear picture about the kind of immune response within the clinical responders (T-helper cell, CTL).

References (73)

  • M. Adams et al.

    Clinical studies of human papilloma vaccines in pre-invasive and invasive cancer

    Vaccine

    (2001)
  • C.J. Hsieh et al.

    Enhancement of vaccinia vaccine potency by linkage of tumor antigen gene to gene encoding calreticulin

    Vaccine

    (2004)
  • A. de Jong et al.

    Enhancement of human papillomavirus (HPV) type 16 E6 and E7-specific T-cell immunity in healthy volunteers through vaccination with TA-CIN, an HPV16 L2E7E6 fusion protein vaccine

    Vaccine

    (2002)
  • W.J. van Driel et al.

    Vaccination with HPV16 peptides of patients with advanced cervical carcinoma: clinical evaluation of a phase I-II trial

    Eur J Cancer

    (1999)
  • G. Meneguzzi et al.

    Immunization against human papillomavirus type 16 tumor cells with recombinant vaccinia viruses expressing E6 and E7

    Virology

    (1991)
  • M.C. Cassetti et al.

    Antitumor efficacy of Venezuelan equine encephalitis virus replicon particles encoding mutated HPV16 E6 and E7 genes

    Vaccine

    (2004)
  • V. Revaz et al.

    Mucosal vaccination with a recombinant Salmonella typhimurium expressing human papillomavirus type 16 (HPV16) L1 virus-like particles (VLPs) or HPV16 VLPs purified from insect cells inhibits the growth of HPV16-expressing tumor cells in mice

    Virology

    (2001)
  • A. Schneider et al.

    Interferon treatment of human genital papillomavirus infection: importance of viral type

    Int J Cancer

    (1987)
  • U.A. Hasan et al.

    TLR9 expression and function is abolished by the cervical cancer-associated human papillomavirus type 16

    J Immunol

    (2007)
  • S.C. Fausch et al.

    Human papillomavirus virus-like particles do not activate Langerhans cells: a possible immune escape mechanism used by human papillomaviruses

    J Immunol

    (2002)
  • K. Matthews et al.

    Depletion of Langerhans cells in human papillomavirus type 16-infected skin is associated with E6-mediated down regulation of E-cadherin

    J Virol

    (2003)
  • M. Gruener et al.

    The E5 protein of the human papillomavirus type 16 down-regulates HLA-I surface expression in calnexin-expressing but not in calnexin-deficient cells

    Virol J

    (2007)
  • H.D. Strickler et al.

    Natural history and possible reactivation of human papillomavirus in human immunodeficiency virus-positive women

    J Natl Cancer Inst

    (2005)
  • M. Scott et al.

    Cell-mediated immune response to human papillomavirus infection

    Clin Diagn Lab Immunol

    (2001)
  • S. Aiba et al.

    Immunohistologic analysis of the phenomenon of spontaneous regression of numerous flat warts

    Cancer

    (1986)
  • N. Coleman et al.

    Immunological events in regressing genital warts

    Am J Clin Pathol

    (1994)
  • I. Arany et al.

    Correlation between mRNA levels of IL-6 and TNF alpha and progression rate in anal squamous epithelial lesions from HIV-positive men

    Anticancer Res

    (2001)
  • H. Hemmi et al.

    Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway

    Nat Immunol

    (2002)
  • D.A. Galloway

    Serological assays for the detection of HPV antibodies

  • I. Jochmus-Kudielka et al.

    Antibodies against the human papillomavirus type 16 early proteins in human sera: correlation of anti-E7 reactivity with cervical cancer

    J Natl Cancer Inst

    (1989)
  • W. Meschede et al.

    Antibodies against early proteins of human papillomaviruses as diagnostic markers for invasive cervical cancer

    J Clin Microbiol

    (1998)
  • M.S. Campo et al.

    Prophylactic and therapeutic vaccination against a mucosal papillomavirus

    J Gen Virol

    (1993)
  • M.C. Feltkamp et al.

    Vaccination with cytotoxic T lymphocyte epitope-containing peptide protects against a tumor induced by human papillomavirus type 16-transformed cells

    Eur J Immunol

    (1993)
  • K. Munger et al.

    Mechanisms of human papillomavirus-induced oncogenesis

    J Virol

    (2004)
  • J.M. Berry et al.

    A review of human papillomavirus vaccines: from basic science to clinical trials

    Front Biosci

    (2003)
  • G.L. Eiben et al.

    Cervical cancer vaccines: recent advances in HPV research

    Viral Immunol

    (2003)
  • Cited by (15)

    • Manipulating IL-10 signalling blockade for better immunotherapy

      2015, Cellular Immunology
      Citation Excerpt :

      Currently, treatment of chronic viral infection and cancer is difficult, therefore, new therapy for chronic viral infection and cancer is urgently needed [18]. Therapeutic vaccination is a potentially promising strategy to control chronically viral infection and cancer [19–25]. It aims at destroying tumour or viral infected cells by inducing robust T cell responses, while leaving the normal cells unharmed.

    • Molecular diagnosis of a laboratory mouse papillomavirus (MusPV)

      2012, Experimental and Molecular Pathology
      Citation Excerpt :

      PVs are non-enveloped small DNA viruses that are highly species-specific, most of which are anatomic site restricted. The species-specificity of HPVs has hindered the development of mouse models, other than transgenic mice, that can reveal clues about HPV carcinogenesis (Campo, 2002; Borzacchiello et al., 2009; Gissmann and Nieto, 2009; Eckert et al., 2000; Stelzer et al., 2010; Kanodia et al., 2010). Other than MusPV (Ingle et al., 2011; Joh et al., 2011), the only PV associated with any mouse species is MmiPV1, isolated from a zoo colony of European harvest mice (Bernard et al., 2010; Sundberg et al., 1996).

    • A prime/boost strategy by DNA/fowlpox recombinants expressing a mutant E7 protein for the immunotherapy of HPV-associated cancers

      2012, Virus Research
      Citation Excerpt :

      Vaccinia virus (VV) recombinants expressing the HPV-16 and HPV-18 E7 proteins have already been used in clinical studies for immunotherapy of cervical cancer and showed high immunogenicity and induction of HPV-specific T-cell-mediated immune responses (Baldwin et al., 2003; Smyth et al., 2004). Clinical trials have also been performed with the attenuated modified vaccinia Ankara (MVA) as a recombinant vector (Acres and Bonnefoy, 2008; Gissmann and Nieto, 2009). However, as MVA replication in mammals is only partially abortive (Blanchard et al., 1998; Drexler et al., 1998), and induces lower immunogenicity than traditional smallpox vaccines (Ferrier-Rembert et al., 2008), the search for new alternatives is always ongoing (Enserink, 2004; Hooper et al., 2007, 2010; Kennedy et al., 2009).

    View all citing articles on Scopus
    View full text