If any field epitomizes the boom and bust cycles of biotech, it would be cancer vaccines. Over the years, numerous tumor immunotherapies have gone through rounds of early-stage successes, only to fail in phase 3 clinical trials. Experts point to many reasons for the failures, from “jumping the gun” before enough was known about the biology or the therapies to letting business considerations—going for low cost and short time lines—trump science; what Peter Bross, chief of clinical evaluations at the US Food and Drug Administration's (FDA's) Center for Biologicals Evaluation and Research calls companies simply not doing their homework. Put these problems together with poorly designed clinical trials of heterogeneous cancer patient populations with late-stage disease, add a lack of familiarity of the regulatory authorities in assessing tumor vaccine products, mix in manufacturing scale-up headaches and the resulting recipe is all but toxic to investors. As Bruce Booth of Atlas Ventures (Waltham, MA, USA) puts it, realizing the potential of cancer vaccines is “full of complexity.”

But some researchers and analysts are keeping the faith, hoping that a more comprehensive understanding of tumor immunology will lead the way to more fruitful approaches (Table 1). Several promising phase 3 programs are nearing completion, so 2009 may well be the year of the cancer vaccine. “There have been other technologies that failed in their first iteration.... As long as modifications are made and something new comes out of it, I think you'll generate interest,” says Reni Benjamin, senior biotech analyst at Rodman and Renshaw (New York).

Table 1 Selected early stage cancer vaccine programs

In the meantime, the question is whether there is enough money to support the approach in the coffers of biotechs or coming from the pharmaceutical industry, which has been burned repeatedly (Table 2). And what lessons from the ever-growing list of failures—and some possible successes—will inform future practitioners in the field?

Table 2 Selected deals in the cancer vaccine sector

Beginnings

Cancer vaccinology is predicated on the notion of awakening the immune system to the presence of cancer by presenting it with antigens associated with tumor cells. Once the immune system is roused, the concept is that it would be capable not only of mounting a sustained bodywide search for similarly suspicious cells, but also of retaining a memory of the abnormal antigens, permitting a renewed, rapid assault should the tumor recur.

The notion that the immune system could be enlisted to launch an attack on an existing tumor has been around at least since the late 1800s, when the New York City–based physician William Coley noticed that metastases at several sites regressed in a sarcoma patient after she developed a bacterial incision-wound infection. Coley's attempts to exploit this discovery were handicapped by the then-crude state of knowledge. But to this day, remnants of this approach can be seen in the use of general immune stimulants, like attenuated bacteria (e.g., mycobacterial components in Bacille Calmette Guerin, BCG) and interleukins, in treating bladder cancer and melanoma, respectively, as well as their inclusion in combination therapies in literally hundreds of clinical trials.

The discovery and identification of tumor-associated antigens, which now number in the hundreds (see Table 3 for some examples), stimulated a second approach to cancer vaccines, an approach still highly visible among the therapies being tested today. Roughly half of ongoing clinical trials enlist a tumor-associated antigen or collection of antigens (Fig. 1 and Table 4). Many such trials have ended in failure, which we now know is because these antigens muster only weak immune responses because they are normal human proteins merely overexpressed on tumor cells (to which the patient would be tolerant) or they too closely resemble such proteins or they elicit only a weak response from the patient's compromised immune system. It is now known that multiple co-stimulatory signals are needed to generate a robust T-cell response against a tumor-associated antigen; if these signals are not supplied, T-cell anergy and peripheral tolerance follows. Such tepid immune responses are not nearly what would be needed to eradicate advanced cancers, which early on accounted for most patients treated in clinical trials. Contemporary trials using tumor-associated and more promising tumor-specific antigens now use various immune stimulatory molecules, such as granulocyte macrophage colony stimulating factor (GM-CSF), and generalized adjuvants, such as keyhole limpet hemocyanin (KLH), to boost the response.

Table 3 Examples of tumor-specific antigens
Figure 1: Cancer vaccine types.
figure 1

(Source: Tufts Center for the Study of Drug Development)

Table 4 Selected cancer vaccines in late clinical trials

Many approaches have explicitly tried to engage cell-mediated immunity either using isolated antigen-presenting cells (APCs) or attempting to stimulate them in situ (Fig. 2). Techniques were developed for extracting dendritic cells, a major APC, loading them up with tumor antigens in various ways and reintroducing them into patients. Early attempts here failed, and in some cases, actually led to poorer outcomes than if the individual had been untreated, as immature dendritic cells, it was later learned, were as likely to suppress the immune system as to stimulate it. Methods for characterizing the right types of dendritic cell and other APCs are now being worked out, and it's become clearer how to activate these cells through cytokines, such as GM-CSF, to optimize antigen presentation (one such immunotherapeutic candidate in late-stage clinical trials, Dendreon's Provenge (Seattle; sipuleucel-T) for prostate cancer, may prove to be among the first therapeutic cancer vaccines to receive FDA approval; Box 1).

Figure 2: Dendritic cells that attack cancer.
figure 2

(Source: National Cancer Institute)

Just in the past five years, information has surfaced, pointing to a whole new problem with cancer immunotherapy—active immunosuppression in the tumor microenvironment. Tumors have been long suspected to evade immune detection by, for example, Darwinian evolution of cells whose defining surface antigens are suppressed or creating positive pressure gradients that make it harder for circulating immune cells to penetrate them (Fig. 3). But now, it has emerged that in addition to evasion, tumors actually can induce local immunosuppression through the stimulation of regulatory T cells or the recruitment of myeloid-derived suppressor (MDS) cells. The former, primarily through their production of transforming growth factor (TGF)-β, inhibit CD8+ cytotoxic T cells (CTLs), T helper 1 (TH1) cells and natural killer (NK) cells, which are the main mediators of immune surveillance against tumors. MDS cells, a mixed population of relatively immature myeloid cells, also suppress cellular immune responses primarily by producing arginase 1 and nitric oxide synthase 2A.

Figure 3: Tumor cell's interactions with the immune system.
figure 3

(Reprinted from Whiteside, T.L. Immune suppression in cancer: effects on immune cells, mechanisms and future therapeutic intervention. Semin. Cancer Biol. 16, 3–15, 2006, with permission from Elsevier.)

One means of potentiating the power of cancer vaccines and unleashing the immune system, according to leading academics, would be to counteract tumor-mediated immune suppression. This could be accomplished by targeting the regulators of the regulators, so to speak. For example, several molecules have been identified (e.g., CTL antigen 4, CTLA-4) that engage with regulatory T cells. Animal studies have shown that blocking such interactions, either with monoclonal antibodies (mAbs) or gene knockouts abrogates immune suppression. Indeed, several dozen clinical trials, according to the US National Institutes of Health (http://www.clinicaltrials.gov), are currently underway using mAbs against CTLA-4 in combination with chemotherapy or vaccines.

Immunotherapy's many faces

Cancer immunotherapy means different things to different people. In the case of cancers that are known to express viral antigens (e.g., cervical cancer and some melanomas that express human papilloma virus), immunotherapy takes the form of a classic immunoprotective, prophylatic vaccine like smallpox or polio where a viral antigen is presented to the immune system. In those cases where cancers overexpress a particular endogenous surface antigen (e.g., Her-2 in some breast cancers or CD-20 in some lymphoma cells), mAbs directed against those surface markers (Genentech's Herceptin (trastuzumab) and Genentech's and Biogen-Idec's Rituxan (rituximab), respectively) provide passive immunity, which can keep a tumor in check for a while. There are many such mAbs for various cancers under development. As currently applied, these mAbs are not preventive but rather therapeutic, though Herceptin has been approved for ever earlier stages in breast cancer, where it might, at least in theory, protect against recurrences by preventing metastases from taking hold.

Active immunotherapies, on the other hand, are designed to incite the individual's own immune system to mount a response to an antigen or group of antigens exclusive to or predominantly associated with the patient's tumor. They can take the form of peptide/protein vaccines or cellular vaccines.

The former type of vaccine generally falls into two categories. The first is based on shared peptide or protein antigens that occur commonly in a particular cancer or group of cancers (epidermal growth factor receptor (EGFR) vIII, for example, which is found in 30–40% of glioblastomas, or MAGE-3, which is expressed on many lung tumors). The proteins can be injected directly or expressed on attenuated virus particles, or nonproliferative bacterial or yeast cells (Box 2). An alternative approach is to isolate antigens from an individual patient and present these back to the person in a form designed to elicit immune surveillance, such as vaccines designed to stimulate responses against antibody idiotypes found on lymphomas or the use of heat shock proteins to present unique tumor peptides (Box 3).

Cellular cancer vaccines can also be divided into two broad groups: allogeneic or autologous. The former, so-called 'off-the-shelf' vaccines, are usually collections of tumor cell lines, administered as aggregates to present several potential tumor antigens to the patient's immune system. Autologous whole cells, on the other hand, are isolated from, and returned to, individuals after some ex vivo manipulation to activate or induce maturation of APCs. An example of this type of vaccine would be a product based on isolation of APCs from a patient that is engineered to express some soluble factor (or factors) that generates an immune response to a common antigen (e.g., prostate-specific antigen in the case of prostate cancer, or p53/telomerase more generally (Box 1)).

Compared with cellular vaccines, peptide vaccines have the advantage of being similar to existing vaccine approaches used for decades in immunization programs against infectious agents. Such vaccines are less tricky to manufacture on a large scale than cellular vaccines. In 2002, for example, the FDA placed a hold on CancerVax's (Carlsbad, CA, USA) phase 3 trial of cellular vaccine Canvaxin because of manufacturing concerns. What's more, the longer clinical history and widespread use of peptide/protein vaccines means that regulators are more familiar with their oversight and less likely to raise issues unanticipated by product sponsors.

The perilous path

Cancer vaccines represent a relatively small portion of the oncology drugs in commercial development. The Tufts Center for the Study of Drug Development (Boston) reports that only one-fifth of oncology biologic therapeutics in company pipelines are vaccines (Fig. 4). Although modern cancer vaccine development dates back to the 1980s, none has been approved in the United States (though there are five products on the market elsewhere; Table 5). Thus, the rate of approval of cancer vaccines lags far behind other biologics—as of 2006, seven of twelve vaccines in phase 3 clinical trials had entered clinical study a decade earlier.

Figure 4: New cancer therapeutics and vaccines entering clinical study per year from 1990 to 2006.
figure 4

(Source: Tufts Center for the Study of Drug Development)

Table 5 Approved and marketed cancer vaccines

To date, an estimated 7,000 people have participated in late-stage clinical trials of active cancer immunotherapies. These have largely been an exercise in frustration, as candidates—including a few that looked quite good in early trials—have fallen by the wayside in pivotal phase 3 trials. Some recent losers that have gone quietly into the night:

  • PANVAC (Therion Biologics, Cambridge, MA, USA), an off-the-shelf vaccine consisting of attenuated poxvirus carrying genes encoding two tumor-associated antigens (carcinoembryonic antigen and mucin 1, MUC-1) and three immunostimulatory molecules (intracellular adhesion molecule 1, B7.1 and lymphocyte function–associated molecule 3) for use in advanced pancreatic cancer, failed to meet clinical endpoints after promising early trials, leading the company to close its doors and file for bankruptcy protection in December 2006.

  • Theratope (Biomira, Edmunton, AB, Canada; now Oncothyreon, Seattle), an off-the-shelf vaccine, consisting of a synthetic mimic (STn-crotyl) of the tumor-associated, O-linked epitope of MUC-1 (STn-serine), tethered to an immunostimulatory protein (KLH) and delivered along with an adjuvant from Seattle-based Corixa (Detox-B, an oil droplet emulsion containing monophosphoryl lipid A and cell wall skeleton from Mycobacterium phlei) for use in metastatic breast cancer, showed no improvement in either time to progression or overall survival. The company hasn't completely abandoned the target; in partnership with Merck KGaA (Darmstadt, Germany), it has developed a “more sophisticated” approach for eliciting a T-cell response, according to Marita Hobman, director of intellectual property management and business development at Oncothyreon.

  • Canvaxin (CancerVax, now MicroMet, Munich), an off-the-shelf mix of three irradiated melanoma cell lines bearing over a dozen defined tumor-associated antigens, plus an adjuvant (BCG) for use in stage III melanoma, yielded worse outcomes in treated patients than in controls, unlike earlier trials in which patients had been more carefully selected for human leukocyte antigen (HLA) alleles correlating with better outcomes. After Canvaxin failed, CancerVax merged with Micromet, which is developing passive immunotherapies using mAbs against various tumor antigens.

  • GVAX (Cell Genesys, S. San Francisco, CA, USA), an off-the-shelf, whole-cell vaccine, consisting of infusions of cells from existing prostate cancer lines engineered to express GM-CSF for use in hormone-refractory prostate cancer, yielded excess deaths in treated patients versus controls, leading to abandonment of the trial.

Although there is a clear preponderance of off-the-shelf vaccines in this group of failures, the fate of individualized vaccines has not necessarily been much better. Two companies with vaccines targeting antibody idiotypes associated with tumors—Favrille (San Diego) and Genitope (Fremont, CA, USA)—both shut down their trials when their products failed to reach statistical significance, essentially ending their programs in late 2008.

Getting it right

A cancer vaccine has to jump through several hoops, says Johns Hopkins University oncologist Hyam Levitsky, co-inventor of GVAX and member of the board of the cancer vaccine company Antigenics (New York). “In an existing tumor, the body has already been exposed to those antigens, so there may already have been an initial immune response. But very often, the immune system is defeated and rendered tolerant to the antigens that the vaccine is targeting. A successful vaccine has to overcome this tolerance, and that's not trivial.” Moreover, Levitsky says, the vaccine frequently has to work in what can be a hostile environment. “The tumor has essentially taken over and altered the landscape, stealing various attributes of the normal immune system to turn down immune response.”

The antigens to use in a vaccine to circumvent the challenge of breaking immune tolerance without generating autoimmunity should be tumor specific. But such antigens are rarely found, says Jeffrey Weber, head of the Comprehensive Melanoma Research Center at the H. Lee Moffitt Cancer Center (Tampa, FL, USA). “These are few and far between. You can discover any number of mutated, tumor-specific antigens, but you seldom find any that turn up on more than 5% of tumors of any given type.” And even when you find one, he says, that doesn't mean it will be highly immunogenic.

In practice, cancer antigens targeted by active immunotherapies have more often been tumor associated: overexpressed on tumors, but nonetheless present at lower frequencies in normal tissues. In trials of vaccines based on these antigens, the necessity of breaking tolerance—for example, by pairing the selected antigen with a powerful adjuvant—has clashed with the need to avoid an excessive immune assault on healthy tissues where the antigen also resides. “You can vaccinate the hell out of somebody against melanoma self-antigens that are overexpressed on cancer, and you won't induce severe side effects—or any immune response to speak of,” says Weber. “But if you administer the same vaccine along with one dose of anti-CTLA-4 antibodies, you can induce life-threatening autoimmune colitis or skin rash or hepatitis.”

Another problem plaguing trials of cancer immunotherapies has been the intractability of the cancers targeted. In theory, any cancer should be amenable to immunotherapy, but in practice, only a few cancers have received most of the attention, at least historically. Melanoma, which early on was found to have tumor-specific antigens, has been targeted frequently using the protein or peptide approach—mostly without success, as no really tumor-specific melanoma antigens have yet been exploited, only tumor-associated antigens. But those cell-based approaches, in which autologous proteins or extracts are used for priming, require access to a sufficient tumor mass. This more or less excludes melanoma or even breast cancer, where the tissue tends to be fibrotic and where tumors tend to be diagnosed increasingly early, while they are still relatively small.

Recognizing that the immune response takes time to develop, some vaccine developers have turned to slow-growing prostate cancer or kidney tumors, where the time to progression is longer. And then, of course, greater prevalence of certain tumor types, such as lung, create a large patient pool with which to populate clinical trials, whereas the dearth of decent treatments for these indications speaks most loudly to the need for ramped-up clinical experimentation.

Certainly, the tendency to use individuals who are in advanced cancer stages has made proof of clinical efficacy more difficult to achieve. Of course, individuals with late-stage disease, who have often been treated with other therapeutic agents that have failed, tend to be more available. And sponsoring companies prefer this population because they expect that positive treatment effects will be observed more quickly in advanced-stage patients than in early-stage or fully resected ones. But decades' worth of clinical trials of cancer vaccines conducted across multiple tumor types not surprisingly suggest that immunotherapies are more likely to work best in patients with earlier-stage, less-aggressive tumors1 or in individuals whose tumor burden has been reduced to the microscopic level by surgery or chemotherapy.

“It's at this level of microscopic disease where I think cancer vaccines are most likely to succeed,” Levitsky says. “Well over 50% of the common cancers can be treated into a state of minimal residual disease. What we lose patients to is typically not the inability to get the disease into that minimal state, but rather the inability to completely eradicate the residual component.” All too often, a seemingly excised tumor returns. “From a public-health point of view,” he says, “the impact of an effective immunotherapy—delivered at the point of minimal residual disease—that could wipe out the last traces of a tumor, would be truly staggering. Ironically, that's probably the most difficult time to demonstrate efficacy in a clinical trial.”

Standard measures of a cancer therapy's efficacy—tumor shrinkage or growth arrest—are worthless for patients with minimal residual disease. How can you score tumor shrinkage if the patient no longer appears to have a tumor? An alternative is to monitor recurrences or, more accurately, deaths among treated versus untreated patients. But that can take a long time. In renal-cell carcinoma, for example, the median time to recurrence for patients who have had their tumors fully resected and show no signs of residual tumor is 6.8 years.

Further complicating cancer immunology trials is the fact that each approach tends to be novel, creating trial-design and regulatory issues. A designated antigen can be either tumor associated or tumor specific, and tumor-specific antigens can be shared by many patients or unique to each patient. Shared antigens offer the prospect of off-the-shelf vaccines, with attendant economies of scale. But they also often lack tumor specificity, thus incurring the drawbacks of immune tolerance.

Whereas the failed Biomira vaccine, Theratope, is an example of a highly purified, well-defined antigen with a single epitope, CancerVax's Canvaxin candidate was a whole-cell mixture with multiple antigens, some of them undoubtedly not even identified, let alone characterized. The former approach runs the risk of eliciting too narrow an immune response. The latter may trigger unwanted cross-reactions, says Weber, and the difficulty of assessing its potency in any given person poses regulatory issues.

The roughly 30 different active cancer immunotherapies now in late-stage clinical trials (Table 3) also differ in their methods of manufacture and the indications for which they're being tested. Trial designs differ greatly, too. Among the variables: early- versus late-stage disease trade-offs, different endpoints and widely divergent timelines due to differential prognoses in different indications.

“Any therapy that's totally novel and first in class is a double-edged sword,” says Mark Frohlich, senior vice president of clinical affairs and chief medical officer of Dendreon. “On the one hand, there's a lot of excitement from patients and regulators who want to approve something that's new and different. On the other hand, if it's a new product, those same regulators also need to make sure they're doing everything to ensure public safety and establish a solid precedent.” Frohlich speaks from experience, having undergone an epic regulatory ordeal with Dendreon's cell-based prostate-cancer vaccine Provenge (Box 1).

For all these reasons, clinical trials of active cancer immunotherapies are high-stakes propositions. Putting a novel approach through its paces among those most likely to benefit—patients who are least ill and for whom obtaining statistically significant results will thus presumably take the most patience—is a costly venture. Even the most sponsor-friendly phase 3 cancer immunotherapy trials—a modest 300-patient study of an easily manufactured, mass-produced off-the-shelf vaccine, in an indication with fast clinical readouts—and associated surgeries, imaging assays and so forth are going to cost about $20 million, according to one knowledgeable company official. With more and larger trials of personalized, more technology-intensive vaccines, the cost soars to hundreds of millions.

The way forward

All segments of the sector—immunologists, entrepreneurs, even regulators—appear to agree that the entrée into cancer vaccines was premature. Thomas Okarma, CEO of Geron (Menlo Park, CA, USA), which has a product in trials, calls early attempts at vaccines “immunological kindergarten.” In addition, certain assumptions about the immune system may not be correct. Early failures with cancer vaccines led to the belief that tumors could not generate an immune response, according to Eli Gilboa, at the University of Miami, Florida. In fact, he says, “It appears [tumors] can [generate an immune response] for a time, but they have elaborated mechanisms for avoiding the immune system. Focusing more on how to mitigate tumor-induced immune suppression will be key going forward.”

A second assumption that is proving false is that chemotherapy and immunotherapy are incompatible. According to Gilboa, evidence is emerging that some forms of chemotherapy are not incompatible, but in fact can synergize immunotherapy.

Another area of agreement is that we have reached the end of the single-agent era. Key to confronting the two main issues facing vaccine developers—the ability of tumors to induce tolerance and the hostile microenvironment in tumors—are combination therapies, but this creates certain problems, according to Robert Schreiber, cancer researcher at Washington University School of Medicine (St. Louis). “Most companies are locked into using their own products and therefore do not like to use combination therapies. And the FDA is particularly leery of trying too many combinations at once,” he says. Schreiber sees a role for university-industry partnerships in getting around this potential logjam. “Once a successful regimen has been identified, there will be many companies that come knocking at the door. Since large-size clinical trials are very expensive, I see a great opportunity for industry and academia/foundations to pair up at this time,” he says.

As for cell-based therapies, the jury is still out on whether autologous vaccines will be the ticket or whether there is a place for allogeneic, off-the-shelf ones. Logistics (read 'cost') seems to dictate that only allogeneic vaccines will be commercially viable, although the evidence to date suggests that it may not be a clinically viable approach.

Even non-cell-based personalized vaccines raise a host of regulatory issues. Is each vaccine a different product? Do vaccines produced for different patients have different immunogenicities? Different cross-reactivities? Different potencies?

The answer might rest in finding more shared, but tumor-specific, antigens, which are in the minority among products in trials today. And down the road, advances in related fields might provide cancer vaccinologists with the tools they need to create off-the-shelf vaccines. For example, Geron, which has an autologous vaccine in trials now, is using this as a proof of concept according to Okarma. The company also has in place the technology for making dendritic cells from stem cells, which would enable the company to prepare an off-the-shelf, activated dendritic cell, something not available at present.

Keith Wonnacott, chief of FDA's cell therapies, joins the chorus of immunologists and academics optimistic that some cancer vaccine will succeed. “We anticipate success, and that lessons will be learned. Much of what has gone on has been helpful. We would love to see success,” he says. Whether that optimism is justified, only time will tell.