NEWS & VIEWS

IMMUNOTHERAPY Both components remain the focus of ques- tions surrounding the optimal design of can- cer (FIG. 1). Numerous pre­clinical vaccines on the move studies in mice have provided ample evidence of the value of cancer vaccines, but clinical Jacques Banchereau and Karolina Palucka translation of this approach, as well as the identification of that elicit thera­peutic The development of therapeutic cancer vaccines has been pursued for many immunity upon , has proved chal- decades. Many vaccines can elicit immunity to tumour antigens, although lenging. The various classes of candidate can- cer antigens include non-mutated proteins their clinical efficacy remains modest. Recent results from two clinical trials against which T‑cell tolerance is incomplete, highlight the potential of personalized vaccination strategies, made possible possibly owing to a tissue-restricted expres- by high-throughput approaches to the identification of immunogenic sion profile that circumvents the mecha- nisms of central and peripheral immune tumour neoantigens. Thus, therapeutic cancer vaccines might soon move tolerance. Other classes comprise peptides into the mainstream. that are entirely absent from the proteome of nonmalignant human cells — either non‑self Refers to Ott, P. A. et al. An immunogenic personal neoantigen for patients with . Nature 547, 5 217–221 (2017) | Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity antigens or neoantigens . Non‑self antigens against cancer. Nature 547, 222–226 (2017) can be derived from viruses, such as human papillomavirus (HPV); such antigens are common between different individuals and, Vaccines against infectious agents — one of the T cells. Only a minority of patients with therefore, a personalized approach to vacci- great successes of medicine — act to prevent cancer respond to this treatment modality3, nation is not required. Nearly a decade ago, . These vaccines, which are designed however, which might reflect — among other vaccination with long, HPV-derived peptides to generate humoral (antibody-based) immu- factors — a low of such T cells. and adjuvants was demonstrated to evoke nity, have mostly been identified in an empiri- Thus, expansion of TAA‑specific T cells, for therapeutic T‑cell immunity in patients with cal manner. By contrast, therapeutic vaccines example, via vaccination, could potentially intraepithelial cervical neoplasia, which was are designed to treat an established , increase the rate of clinical responses to correlated with clearance of HPV and regres- such as cancer, mainly by evoking cellular immune-checkpoint inhibitors. sion of neoplastic lesions6. Neoantigens are (T‑cell-based) immune responses. The first Vaccines are composed of antigens and created by cancer-specific DNA alterations attempts to vaccinate humans against can- adjuvants — that is, activators of -­ that result in unique peptide sequences5. cer were made more than a century ago1. presenting cells that shape immune responses. The identification of these neoantigens has In the 1950s, studies in mice revealed that chemically-induced­ tumours were immuno- Molecularly-defined genic and could elicit therapeutic immunity. non-self antigens and/or Since then, numerous teams have worked on neoantigens • Viral antigens defining the mechanisms of these responses, • High mutational load: with Robert Schreiber propounding the Whole-tumour preparations mutated peptides now prominent ‘three Es’ concept of cancer A mixture of undefined self Purified self-antigens • Low mutational load: immunity and immunoediting: elimination, antigens, non-self antigens, Overexpressed or aberrantly subclonal antigens equilibrium, and escape2. The discovery and neoantigens expressed tissue antigens • Novel forms to be discovered of co‑stimulatory and co‑inhibitory path- ways controlling the extent of the led to the development of immune-­ Polyclonal immune response Need to break immune Focused immune response checkpoint inhibitors, such as anti‑PD‑1 with random expansion tolerance of self-antigens for with high frequency of antibodies, which can counter the ‘escape’ of of neoantigen-specific T cells anticancer response neoantigen-specific T cells from immunity3,4. Important roles Figure 1 | Generations of antigens. Early clinical studies used whole cancer Nature Reviews | Clinical of immune-checkpoint inhibitors in anti­ cell preparations (for example, tumour lysates) as the source of tumour antigens, comprising a cancer have already been established; mixture of self-antigens and undefined neoantigens. These vaccines elicited broad immune their clinical effect, which can be durable responses, possibly not reaching a critical threshold for the frequency of anticancer T cells across various cancers, is generally associated needed for clinical efficacy. The next stage of cancer vaccine development was focused on puri- with a high mutational load, which might, fied self-antigens, the use of which requires breaking of T‑cell tolerance. Most recently, defined in turn, be associated with the presence of neoantigens have been pursued as drivers of a focused immune response that enables the tumour-associated antigen (TAA)-specific critical thresholds for the frequency of anticancer T cells and clinical efficacy to be reached.

NATURE REVIEWS | CLINICAL ONCOLOGY www.nature.com/nrclinonc ©2017 Mac millan Publishers Li mited, part of Spri nger Nature. All ri ghts reserved.

NEWS & VIEWS

been made possible by the advent of next-­ linker-connected 27‑amino-acid (27mer) enrichment of otherwise poorly represented generation sequencing (NGS), which ena- peptides, corresponding to 10 selected neo- neoantigens. Under these conditions, the lim- bles efficient sequencing of both RNA and antigens per patient (and five in one patient), ited choice of mutated peptides will require DNA from cancers, and the development of were delivered via injection into the in­­ the development of highly immunogenic vac- novel algorithms for predicting the binding guinal lymph nodes. After eight doses, T‑cell cines to prime a new T‑cell repertoire. Thus, of peptides to MHC class I and class II pro- responses against 60% of the 125 mutations our improved understanding of the func- teins. In general, neoantigens are unique to represented in the poly‑neoepitope vac- tions of dendritic cells might prove essential, each patient, although patients deficient in cines were detected; each patient developed by enabling the development of cell-based mismatch-­repair enzymes have exceptionally an immune response against at least three vaccines that elicit an immune response to high numbers of common somatic mutations, mutations. Eight patients who had no visible rare mutations that is sufficient to induce a which seem to predict a therapeutic response tumours at the time of vaccination remained novel repertoire of efficient effector T cells. to anti‑PD‑1 antibodies across different can- disease-free 12–23 months later. The remain- We are confident that the coming years will cers7. Some tumours, including most mela­ ing five patients had relapses and progressive witness the emergence of cancer vaccines as a nomas, have high mutation rates, but whether disease at the time of vaccination; notably, major modality for the management of cancer. this is reflected in the generation of neo­ in one of these patients, combined treat- Through a logical, evidence-based approach, antigens remains to be established. Indeed, ment with an anti‑PD‑1 antibody resulted building on the immense amount of know­ certain criteria need to be met for mutations in a complete response9. The results of these ledge that has been generated in immun­ology to create neoantigens: they must be present two studies confirm the over the past 50 years, what started more within the peptides that are processed by the of melanoma neoantigens, as observed in a than 100 years ago with empirical findings antigen-presentation machinery; the mutated previous proof‑of‑concept clinical study of related to therapeutic cancer vaccines will be peptide must bind to MHC proteins with suf- a dendritic-cell vaccine10. transformed into a major instrument of the ficient affinity to be presented to T cells; and So, what it is the next step for the develop­ therapeutic armamentarium. the MHC-bound mutated peptide must be ment of cancer vaccines? Randomized Jacques Banchereau and Karolina Palucka are at recognized by the patient’s T cells. phase II studies with larger cohorts of The Jackson Laboratory for Genomic Medicine, Two recent phase I studies illustrate the patients will be necessary to assess the actual 10 Discovery Drive, Farmington, potential of personalized cancer vaccines therapeutic activity of these vaccines. Studies Connecticut 06032, USA. targeting neoantigens identified using are also needed to optimize the nature of [email protected]; NGS and predictive MHC-binding algo- the vaccine components, assess the relative [email protected] rithms8,9. Ott et al.8 enrolled 10 patients with advantage of peptide versus RNA-based doi:10.1038/nrclinonc.2017.149 stage IIIB/C or IVM1a/b melanoma in order approaches, and to develop combination Published online 12 Sep 2017 to evaluate a personalized peptide-based , evaluate their dosing, and ulti- 1. Currie, G. A. Eighty years of immunotherapy: a review vaccination approach. The tumours from mately their timing. Future immunological of immunological methods used for the treatment of eight of the 10 patients had a high muta- studies will refine the mechanisms of vac- human cancer. Br. J. Cancer 26, 141–153 (1972). 2. Dunn, G. P., Old, L. J. & Schreiber, R. D. The three Es tion rate; for these eight patients, 13–20 cine responses and their clinical efficacy. In of cancer immunoediting. Annu. Rev. Immunol. 22, neoantigens predicted to be presented on the two clinical trials8,9, most of the responses 329–360 (2004). + + 3. Sharma, P. & Allison, J. P. The future of immune tumour cells were synthesized as immu- detected involved CD4 , rather than CD8 , checkpoint therapy. Science 348, 56–61 (2015). nizing peptides containing 15–30 amino T cells, underscoring the need for further 4. Lesokhin, A. M., Callahan, M. K., Postow, M. A. & Wolchok, J. D. On being less tolerant: enhanced acids. Six of these patients underwent studies to uncover the location of specific cancer immunosurveillance enabled by targeting vaccination with these personalized neo­ T‑cell subsets (blood versus tumour ver- checkpoints and agonists of activation. Sci. Transl Med. 7, 280sr1 (2015). antigen peptides, together with the adjuvant sus lymph nodes), as well as their function 5. Schumacher, T. N. & Schreiber, R. D. Neoantigens in poly‑ICLC, beginning a median of 18 weeks in the control of cancer. Furthermore, whether . Science 348, 69–74 (2015). 6. Kenter, G. G. et al. Vaccination against HPV‑16 after tumour resection with curative intent. the observed responses are attributable to oncoproteins for vulvar intraepithelial neoplasia. Vaccine-induced polyfunctional CD4+ and reactivation of memory T cells or priming N. Engl. J. Med. 361, 1838–1847 (2009). + 7. Le, D. T. et al. Mismatch repair deficiency predicts CD8 T cells were found to target 58 (60%) of naive T cells is not known; even when an response of solid tumors to PD‑1 blockade. and 15 (16%) of the 97 unique neoantigens anticancer immune response is undetectable Science 357, 409–413 (2017). 8. Ott, P. A. et al. An immunogenic personal neoantigen used across all patients, respectively. At in baseline blood samples, memory T cells vaccine for patients with melanoma. Nature 547, 20–32 months after vaccination, the four might be present at frequencies below the 217–221 (2017). 9. Sahin, U. et al. Personalized RNA mutanome vaccines patients with stage III disease were recur- detection threshold, and/or might reside mobilize poly-specific therapeutic immunity against rence‑free. Two patients with lung meta­ in other tissues. A critical question that is cancer. Nature 547, 222–226 (2017). 10. Carreno, B. M. et al. Cancer immunotherapy. A stases had disease recurrence shortly after at the core of this approach, and remains vaccine increases the breadth and the last vaccination, but both had a complete to be solved, is which antigens should be diversity of melanoma neoantigen-specific T cells. Science 348 response to subsequent treatment with the targeted in tumours with a low mutational , 803–808 (2015). anti‑PD‑1 antibody pembrolizumab, with burden and, possibly, a low frequency of Acknowledgements We thank the patients and healthy volunteers who, over the evidence of expansion of their neo­antigen- cancer-­specific memory T cells that can be years, have agreed to participate in our studies. The work of specific T‑cell repertoires8. In the second reinvigorated. Immunologically dormant the authors is supported, in part, by grants from the US NIH 9 (5UO1 AI124297 and 5RO1 AG052608 to J.B.; study, Sahin et al. enrolled 13 patients with neoantigens might be uncovered, or possibly 5R01 CA195712 to K.P.), and from the US Department of stage III or IV melanoma, and used person- induced, during , radiotherapy, Defense (W81XWH‑17‑1‑0010 to K.P.). alized RNA‑based ‘poly-epitope’ vaccines: and/or , with the clonal selec- Competing interests statement two synthetic RNAs, each encoding five tion of cancer cells potentially facilitating The authors declare no competing interests.

NATURE REVIEWS | CLINICAL ONCOLOGY www.nature.com/nrclinonc ©2017 Mac millan Publishers Li mited, part of Spri nger Nature. All ri ghts reserved.