Commentary (Fox)-Melanoma Vaccines: What We Know So Far

Publication
Article
OncologyONCOLOGY Vol 19 No 1
Volume 19
Issue 1

Drs. Bystryn and Reynoldspresent an overview of melanomavaccines, including atheoretical rationale to support the approach,criteria for an effective vaccine,and a discussion of the challengesto optimal vaccine design. Results ofclinical trials where vaccine-inducedimmune responses correlated withimproved clinical outcome are discussed,as well as limitations of monitoringvaccine-induced immuneresponses. A series of randomized, concurrentlycontrolled trials with complex,polyvalent whole-cell vaccines,extracts, lysates, or shed antigens arereviewed. The authors conclude thatmelanoma vaccines' "potentially mostsignificant application" may be the preventionof melanoma in individuals athigh risk of developing the disease.Their review discusses the generallyaccepted rationale for selecting vaccineantigens and does a thoughtfuljob of reviewing the current state ofcomplex melanoma vaccines.

Drs. Bystryn and Reynolds present an overview of melanoma vaccines, including a theoretical rationale to support the approach, criteria for an effective vaccine, and a discussion of the challenges to optimal vaccine design. Results of clinical trials where vaccine-induced immune responses correlated with improved clinical outcome are discussed, as well as limitations of monitoring vaccine-induced immune responses. A series of randomized, concurrently controlled trials with complex, polyvalent whole-cell vaccines, extracts, lysates, or shed antigens are reviewed. The authors conclude that melanoma vaccines' "potentially most significant application" may be the prevention of melanoma in individuals at high risk of developing the disease. Their review discusses the generally accepted rationale for selecting vaccine antigens and does a thoughtful job of reviewing the current state of complex melanoma vaccines.

Vaccine Shortcomings

My own perspective is that while such complex vaccines are the "correct" antigen source, these vaccines will not reach their therapeutic potential, because, alone, they cannot overcome tolerance or tumor-induced suppression. The authors describe preclinical studies in which whole-tumor vaccines protected naive animals from a subsequent lethal tumor challenge. However, at the beginning of 2005 there is essentially no evidence that therapeutic vaccination alone can cure animals with large established (7- to 10-day old) poorly immunogenic tumors. Why does vaccination work in naive "tumor-free" animals but not in animals with established tumor burden? While tumors have the capacity to directly or indirectly suppress the antitumor immune response, this is only part of the answer.

What is needed for immune-mediated tumor destruction? As noted by Bystryn and Reynolds, preclinical models generally document that CD8 T cells are the primary mediators of a curative immune response. However, it is becoming increasingly clear that CD4+ T cells are essential for longterm persistence of tumor-specific CD8 T cells and "cure" of animals with established tumors.[1] While many vaccine studies report a significant increase in the frequency of tumor- antigen-specific T cells, these frequencies are generally below 1% of the total T-cell population. Our Institute's view, supported by preclinical data and recent clinical reports, is that this level of response will not provide a curative effect, explaining why melanoma vaccine strategies have not delivered on their promise.[2]

Improving Vaccine Therapy

What can be done to overcome these tumor-induced roadblocks and increase the frequency of tumor-specific T cells in melanoma patients?

(1) Use complex antigen vaccines: While class I-presented peptide vaccines have provided proof of concept and opportunities for sophisticated immunologic monitoring, vaccines utilizing CD8 epitopes alone should be broadened to include CD4 epitopes or switched to complex vaccines. Immune responses composed of CD4 and CD8 T cells against a spectrum of tumor antigens will be required to maintain long-term curative immunity.

(2) Provide costimulation: At least three costimulation approaches are in or nearing the clinic. The most clinically advanced is anti-CTLA-4, which interferes with a natural "braking" system that limits costimulation. While serious autoimmune reactions have been noted following anti-CTLA-4 administration, they have been controllable and their detection is seen as evidence of a fundamental change in the nature of the induced immune response.[ 3] Additional antibodies that provide costimulatory signals and directly augment immune cell function are anti-4-1BB and anti-OX-40. Our Institute has developed an anti-OX40 antibody that should enter clinical trials in 2005.

(3) Block suppression: Antibodies, small molecules and/or receptor antagonists that interfere with these suppressive cytokines and molecules are becoming available and should soon find their way into cancer vaccine trials.

(4) Eliminate "suppressor" regulatory T cells (Treg) cells: Since Treg cells express interleukin (IL)-2 receptors, investigators are attempting to eliminate them, using an IL-2-toxin fusion protein (Ontak). In the near future, we expect that reagents will be available to selectively deplete and/or transiently regulate these cells or their function.

(5) Restructure the "environment" for immune cells: Manipulating costimulation, suppressive molecules and/or Treg cells may augment the immune response to vaccination, but homeostatic mechanisms may limit the increase in tumor-specific T cells. Building on the findings of Mackall and colleagues,[4] our Institute has developed a strategy that may overcome the homeostatic controls that maintain a fixed number of lymphocytes. In preclinical models, this strategy-which involves vaccinating a lymphopenic mouse that has been reconstituted with 1% to 10% of their normal lymphocytes-results in higher frequencies of tumor-specific T cells and increased therapeutic effects in vaccine and adoptive transfer models.[5,6]

Ongoing Research

In collaboration with colleagues in Xi'an, China (Drs. J. Ma, Y. Wang, and colleagues at Xi'an Jiaotong University, studying ovarian cancer) and Munich, Germany (Drs. D. Ruttinger, M. Schlemer, R. Hatz, and colleagues at Klinikum Grosshadern, studying non-small-cell lung cancer), our Institute is participating in four planned clinical trials of this reconstituted lymphopenic patient (RLP) approach.[7] These investigations include a trial for HLA-A2+ metastatic melanoma patients (Drs. W. J. Urba, B. Curti, and H. Ross, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon). The RLP trial design involves the processing and storage of an apheresis product, administration of chemotherapy to induce lymphopenia, infusion of the apheresis product, and vaccination.[7]

Conclusions

Ultimately, the "success" of melanoma vaccines will require a multifactorial approach, which will not be easy to establish. Combining new agents from different companies will result in a need to deal with intellectual property issues as well as corporate concerns regarding possible adverse events when agents are combined. Success in the area will require a positive perspective and collegial relationships between corporate, academic, funding, and regulatory agencies. The Cancer Vaccine Consortium of the Sabin Vaccine Institute recently initiated a series of working groups to address these and other issues, in an effort to facilitate development of "cutting edge" vaccine trials that combine multiple agents and may increase objective clinical response rates.

Another roadblock to cancer vaccine development is the paucity of translational investigators, specifically, scientists who are actively involved in translating their basic science observations to the clinic. In my opinion, this is a crisis that severely limits the application of new technology to the clinic. Recognizing this, the International Society for Biological Therapy of Cancer (iSBT) is working to increase participation by PhD scientists and students. However, solving this crisis will require the institution of translational medicine training programs that develop scientists interested in transferring good basic science to patient care and the establishment of mechanisms to recognize, support, and reward scientists that pursue this career path.

Financial Disclosure:The author is a member of the scientific advisory board for Cell Genesys Inc.

Disclosures:

The author is a member of the scientific advisory board for Cell Genesys Inc.

References:

1. Hu HM, Winter H, Urba WJ, et al: Divergent roles for CD4+ T cells in the priming and effector/memory phases of adoptive immunotherapy. J Immunol 165:4246, 2000.
2. Rosenberg SA, Yang JC, Restifo NP: Cancer immunotherapy: Moving beyond current vaccines. Nat Med 10:909, 2004.
3. Robinson MR, Chan CC, Yang JC, et al: Cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma: A new cause of uveitis. J Immunother 27:478, 2004.
4. Mackall CL, Bare CV, Granger LA, et al: Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing. J Immunol 156:4609, 1996.
5. Ma J, Urba WJ, Si L, et al: Anti-tumor T cell response and protective immunity in mice that received sublethal irradiation and immune reconstitution. Eur J Immunol 33:2123, 2003.
6. Hu HM, Poehlein CH, Urba WJ, et al: Development of antitumor immune responses in reconstituted lymphopenic hosts. Cancer Res 62:3914, 2002.
7. Ma J, Poehlein CH, Jensen S, et al: Manipulating the host response to autologous tumor vaccines. Dev Biol 116:93, 2004.

Related Videos
Interim data reveal favorable responses in patients with low-grade serous ovarian cancer treated with avutometinib plus defactinib, according to Susana N. Banerjee, MD.
Treatment with mirvetuximab soravtansine appears to produce a 3-fold improvement in objective response rate vs chemotherapy among patients with folate receptor-α–expressing, platinum-resistant ovarian cancer in the phase 3 MIRASOL trial.
PRGN-3005 autologous UltraCAR-T cells appear well-tolerated and decreases tumor burden in a population of patients with advanced platinum-resistant ovarian cancer.
An expert from Dana-Farber Cancer Institute discusses findings from the final overall survival analysis of the phase 3 ENGOT-OV16/NOVA trial.