Does patient-tailored immunotherapy pave the way for new renal cell carcinoma treatment perspectives?
Editorial

Does patient-tailored immunotherapy pave the way for new renal cell carcinoma treatment perspectives?

Ilse Van Brussel1, Amaryllis H. Van Craenenbroeck2, Dorien M. Schrijvers3, Nathalie Cools4

1Laboratory of Pharmacology, University of Antwerp, B-2610 Antwerp, Belgium; 2Division of Nephrology-Hypertension, Antwerp University Hospital, B-2650 Edegem, Antwerp, Belgium; 3Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium; 4Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Antwerp, Belgium

Correspondence to: Ilse Van Brussel. Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium. Email: ilse.vanbrussel@ua.ac.be.

Submitted Sep 10, 2012. Accepted for publication Oct 11, 2012.

doi: 10.3978/j.issn.2223-4683.2012.10.02


Renal cell carcinoma (RCC) constitutes 80 to 85 percent of primary renal neoplasms in adults (1). Although surgical resection can be curative in localized disease, many patients eventually recur. Patients with locally advanced or metastatic disease have a poor prognosis, with a 5-year survival rate of less than 15%. Because RCC is highly resistant to both chemotherapy and radiation therapy (2), immunotherapy might have the highest potential as a novel treatment for RCC (3). Accumulating evidence indeed shows that effector cells of the immune system play an important role in the recognition and elimination of neoplastic cells. Particularly in RCC, experimental data suggest that the immune system might be extremely important in controlling the disease in situ (4). For example, in RCC, major T cell infiltrates can be demonstrated in the tumor. The presence of antigen-specific T cell clones has been shown in both primary lesions and draining lymph nodes, and these clones are able to lyse renal carcinoma cells in vitro (5,6). Unfortunately, in the long term, the immune response against RCC fails to limit disease progression.

To date, several immunotherapeutic approaches have been proposed to treat RCC (3,7), particularly since different RCC-related tumor antigens have been identified that can be targeted, processed and presented by immune effector cells (8). It is well known that of these immune effector cells, dendritic cells (DCs) play an orchestrating role in regulating T cell responses, partly due to their potent antigen-presenting capacity (9). The attractive concept of autologous monocyte-derived DC-based tumor vaccination resulted in an increasing number of phase I/II trials with different approaches regarding the vaccine composition, including the nature of the antigen(s) (7). Synthetic peptides are commonly used to load DCs in DC-based vaccination trials, but are mostly HLA-A*0201-restricted which limits their clinical use. The use of tumor lysate circumvents this restriction and has the advantage of inducing a polyclonal immune response. Similarly, the use of total renal tumor RNA-transfected DCs has proven to induce T cell activities directed against a broad set of renal tumor-associated antigens (10). In the AGS-003 strategy, autologous DCs co-electroporated with the patients’ amplified tumor mRNA and synthetic CD40L RNA are employed (11). Although encouraging results are reported upon administration of this vaccine with regard to immune response as well as survival (12), results from the phase III trial have not been published yet (http://clinicaltrials.gov/ct2/show/NCT01582672).

Recently, the first autologous DC-based therapy was approved by the Food and Drug Administration (FDA) for treatment of patients with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). Sipuleucel-T (known by the trade name, “Provenge”) is a cellular vaccine which is created upon collection of patient’s white blood cells and subsequent in vitro incubation of these cells with a fusion protein that combines prostate acid phosphatase (PAP) with recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF). Upon re-infusion of this cell product into the patient, sipuleucel-T stimulates the patient’s own immune system to specifically recognize and combat his cancer. As was published in the New England Journal of Medicine, sipuleucel-T prolonged median survival by 4.1 months compared with results in placebo-treated patients (13). In summary, the advantage of DC-based immunotherapeutic strategies is good tolerability and observed survival benefit. Unfortunately, these patient-tailored therapeutics are very time-consuming and costly. As an alternative, targeting DCs in vivo may be more attractive from a cost-effectiveness perspective, since this approach would omit tailor-made ex vivo culturing. Indeed, such off-the-shelf therapeutic vaccines have shown preliminary evidence of efficacy (13), providing hope that improvements in patient outcomes with this modality may lead to therapeutic options that are less resource-intense. Walter et al. (14) developed IMA901, a peptide vaccine for RCC, consisting of nine HLA-A*02-restricted tumor-associated peptides (TUMAPs) and one HLA-DR-restricted TUMAP in combination with administration of GM-CSF. GM-CSF is used to stimulate antigen-presenting cells (APCs), including DCs (15), in vivo. Subsequently, TUMAPs bind to major histocompatibility complex (MHC) molecules on the cell surface of APCs, and in turn the activated APCs facilitate in vivo priming of T cells. Hence, the designated purpose of IMA901 is to elicit a therapeutic immune response to antigens expressed by cancer cells. The authors report stabilization of the disease or a partial response to therapy in 43% of the 28 patients that received eight intradermal IMA901 vaccinations. In the remainder 57% of subjects, RCC progressed (14). Furthermore, subjects that responded to multiple TUMAPs were significantly more likely to experience disease control than subjects that responded to only one TUMAP or showed no response (14), indicating that the enhancement of the breadth of immune responses targeted to antigens introduced by the vaccine is of great consequence.

Strong and broad T cell responses will prevent immune escape by certain cancer cells that have altered their cell surface expression of certain HLA molecule(s). Although Walter et al. have not addressed the added value of inclusion of the HLA-DR-restricted TUMAP, others have demonstrated the importance of CD4+ T cell help in the stimulation of such strong and effective cellular immune responses. CD4+ T helper cells deliver help for CD8+ cytotoxic T cells by fully activating DCs through the CD40-CD40 ligand signaling pathway as well as by the secretion of interleukin-2 (16). Pan HLA-DR epitope (PADRE) peptides, that are capable of binding to different MHC class II molecules with high-affinity (17), have been used in conjunction with other forms of vaccines to enhance vaccine potency in preclinical models (18,19) and they have also been used in clinical trials with minimal toxicity (20). Alternatively, CD4+ T cell help can be achieved by using synthetic long peptides (SLPs) (21). Following in vivo uptake by DCs, a proportion of the SLPs is processed and loaded into MHC class II molecules, allowing fragment presentation to CD4+ T helper cells. Another part of the ingested SLPs is digested by the proteasome in the cytosol and the endoplasmatic reticulum. This is followed by loading of 8-10 amino acid-long peptides into MHC class I molecules, which allows fragment presentation to CD8+ cytotoxic T cells (22).

Nevertheless, the increase in median overall survival in the patients treated with IMA901 was not associated with standard measures of efficacy, including changes in size and volume of measurable lesions. This uncoupling effect on survival and disease progression appears to be a common property of immunotherapy, and is designated as a delayed treatment effect. Indeed, biological effects of cancer vaccines are not related to their pharmacokinetics, and effectiveness may take weeks or months to become apparent (23). Hence, effectiveness as measured by tumor regression at traditionally early time points may fail to demonstrate any measurable potentially beneficial effect. For this, studies are intensified to develop new, non-invasive diagnostic tests, e.g., biomarkers, to carefully monitor the effect of the vaccination strategy on the tumor. The feasibility and value of a comprehensive biomarker program has been underscored by Walter et al., as indicated by the identification of two biomarkers, APOA1 and CCL17, that are potentially predictive for vaccine-induced immunity and overall survival.

Furthermore, studies that improve or refine immunotherapeutic outcomes in the clinic are warranted. Simple methods that are likely to increase efficacy include (I) administration of boost vaccinations in order to extend the response; (II) treating patients earlier in their disease course; (III) and combination strategies with agents that are known to activate, accelerate, and augment immune responses (24). These include adjuvants (IL-7, IL-12, IL-15, and monophosphoryl lipid) to augment T cell responses, and antagonists of negative regulators of T cell activation [anti-CTL-associated receptor 4 (CTLA-4) and anti-programmed death 1 (PD1) receptors], as well as agents to neutralize immunosuppressive cytokines (anti-IL-10 and anti-TGF-β) that are important in winding down immune responses. Walter and colleagues (14) improved the efficacy of the IMA901 vaccination by means of a single-dose cyclophosphamide. Indeed, the median overall survival of the patients treated with IMA901 and cyclophosphamide was 23.5 months compared to 14.8 months in the patients treated without cyclophosphamide. It has been postulated that this immunomodulator counteracts the regulatory mechanisms that oppose successful immunotherapy, e.g., by reducing the numbers of regulatory T cells (Tregs) (25).

In conclusion, it remains to be established from ongoing phase III trials whether the DC-based vaccine AGS-003 or the peptide vaccine IMA901 results in the best treatment for advanced renal cell carcinoma with the highest overall survival benefit. However, it is likely that vaccination approaches will become part of the armamentarium of nephrologists, urologists and medical oncologists who manage and care for renal cancer patients.


Acknowledgements

None.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Chow WH, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nat Rev Urol 2010;7:245-57. [PubMed]
  2. Ferguson RE, Jackson SM, Stanley AJ, et al. Intrinsic chemotherapy resistance to the tubulin-binding antimitotic agents in renal cell carcinoma. Int J Cancer 2005;115:155-63. [PubMed]
  3. Itsumi M, Tatsugami K. Immunotherapy for renal cell carcinoma. Clin Dev Immunol 2010;2010:284581.
  4. Michael A, Pandha HS. Renal-cell carcinoma: tumour markers, T-cell epitopes, and potential for new therapies. Lancet Oncol 2003;4:215-23. [PubMed]
  5. Van den Hove LE, Van Gool SW, Van Poppel H, et al. Phenotype, cytokine production and cytolytic capacity of fresh (uncultured) tumour-infiltrating T lymphocytes in human renal cell carcinoma. Clin Exp Immunol 1997;109:501-9. [PubMed]
  6. Caignard A, Guillard M, Gaudin C, et al. In situ demonstration of renal-cell-carcinoma-specific T-cell clones. Int J Cancer 1996;66:564-70. [PubMed]
  7. Draube A, Klein-González N, Mattheus S, et al. Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and meta-analysis. PLoS One 2011;6:e18801. [PubMed]
  8. Finke JH, Salvucci-Kierstead L, Ranieri E, et al. Immunologic response to renal cell carcinoma. In: Bukowski RM, Novick AC. eds. Renal cell carcinoma: molecular biology, immunology and clinical management. Totowa, NJ (USA): Humana Press, 2000:39-62.
  9. Cools N, Ponsaerts P, Van Tendeloo VF, et al. Balancing between immunity and tolerance: an interplay between dendritic cells, regulatory T cells, and effector T cells. J Leukoc Biol 2007;82:1365-74. [PubMed]
  10. Su Z, Dannull J, Heiser A, et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res 2003;63:2127-33. [PubMed]
  11. Figlin RA, Amin A, Dudek A, et al. Phase II study combining personalized dendritic cell (DC)-based therapy, AGS-003, with sunitinib in metastatic renal cell carcinoma (mRCC). J Clin Oncol 2012;30:abstr 348.
  12. Healey D, Gamble AH, Amin A, et al. Immunomonitoring of a phase I/II study of AGS-003, a dendritic cell immunotherapeutic, as first-line treatment for metastatic renal cell carcinoma. J Clin Oncol 2012;28:abstr e13006.
  13. Kantoff PW, Schuetz TJ, Blumenstein BA, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol 2010;28:1099-105. [PubMed]
  14. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012;18:1254-61. [PubMed]
  15. Markowicz S, Engleman EG. Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of human peripheral blood dendritic cells in vitro. J Clin Invest 1990;85:955-61. [PubMed]
  16. Schuurhuis DH, Laban S, Toes RE, et al. Immature dendritic cells acquire CD8(+) cytotoxic T lymphocyte priming capacity upon activation by T helper cell-independent or -dependent stimuli. J Exp Med 2000;192:145-50. [PubMed]
  17. Alexander J, Sidney J, Southwood S, et al. Development of high potency universal DR-restricted helper epitopes by modification of high affinity DR-blocking peptides. Immunity 1994;1:751-61. [PubMed]
  18. Wierecky J, Müller MR, Wirths S, et al. Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Res 2006;66:5910-8. [PubMed]
  19. Alexander J, del Guercio MF, Frame B, et al. Development of experimental carbohydrate-conjugate vaccines composed of Streptococcus pneumoniae capsular polysaccharides and the universal helper T-lymphocyte epitope (PADRE). Vaccine 2004;22:2362-7. [PubMed]
  20. Kavanagh B, Ko A, Venook A, et al. Vaccination of metastatic colorectal cancer patients with matured dendritic cells loaded with multiple major histocompatibility complex class I peptides. J Immunother 2007;30:762-72. [PubMed]
  21. Kenter GG, Welters MJ, Valentijn AR, et al. Phase I immunotherapeutic trial with long peptides spanning the E6 and E7 sequences of high-risk human papillomavirus 16 in end-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res 2008;14:169-77. [PubMed]
  22. Melief CJ, van der Burg SH. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat Rev Cancer 2008;8:351-60. [PubMed]
  23. Hoos A, Eggermont AM, Janetzki S, et al. Improved endpoints for cancer immunotherapy trials. J Natl Cancer Inst 2010;102:1388-97. [PubMed]
  24. Van Brussel I, Berneman ZN, Cools N. Optimizing dendritic cell-based immunotherapy: tackling the complexity of different arms of the immune system. Mediators Inflamm 2012;2012:690643.
  25. Yao X, Ahmadzadeh M, Lu YC, et al. Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 2012;119:5688-96. [PubMed]
Cite this article as: Van Brussel I, Van Craenenbroeck AH, Schrijvers DM, Cools N. Does patient-tailored immunotherapy pave the way for new renal cell carcinoma treatment perspectives? Transl Androl Urol 2013;2(2):85-88. doi: 10.3978/j.issn.2223-4683.2012.10.02

Download Citation