Guest guest Posted December 18, 2007 Report Share Posted December 18, 2007 This is a little scary after ya'll talking about that movie Crystal ________________________________________________________________________________\ _ Tissue Vaccines for Cancer Mark A Suckow; Heinrich; Elliot D Rosen Expert Rev Vaccines. 2007;6(6):925-937. ©2007 Future Drugs Ltd. Posted 12/13/2007 Abstract and Introduction Abstract Most tumors, including prostate carcinoma, are heterogeneous mixtures of neoplastic cells and supporting stromal matrix. Attempts to vaccinate as a means to treat or prevent cancer have typically relied on use of a single antigen or cell type. In the case of whole-cell vaccines, clonal populations of cancer cells are grown in culture and harvested for vaccine material. However, it is clear from microarray data that neoplastic cells grown in culture are greatly different from those found in vivo. Tissue vaccines are harvested directly from tumors and are used to immunize the animal or the patient. They are antigenically rich, in that they are comprised of not only neoplastic cells but also supporting stromal matrix; furthermore, they include antigens that may be expressed only in vivo and which may be critical to a successful immune response to the cancer. For these reasons, the idea that tissue vaccines for cancer have potentially great utility has merit and should be explored further. Introduction Cancer is a group of diseases characterized by uncontrolled growth and spread of abnormal cells. The clinical outcome of cancer is often significant debilitation or death. Although great strides have been made in the clinical approach to cancer, the disease remains a substantial challenge to clinicians and scientists alike. It is projected that 1,444,920 new cancer cases will be diagnosed and 559,650 people will die from cancer in the USA in 2007.[1] In this paper, prostate cancer is given particular focus as a solid tissue tumor exemplary of many other tumor types. Cancer of the prostate gland is the most commonly diagnosed cancer in men and the second most common cancer resulting in death of men, on an age-adjusted basis.[1] In the majority of cases, prostate carcinoma is a disease of older men, many of whom have comorbid conditions that elevate the risk of cancer death.[2-10] Although the precise mechanisms that lead to the development of prostate cancer are yet to be defined, it is evident that prostate cancer develops as the sum result of genetic and epigenetic changes, including those that inactivate tumor-suppressor genes and activate oncogenes.[11,12] For example, the inheritance of multiple genetic factors in humans, such as ELAC2; a gene in the HPC1 region encoding for 2'-5'-oligoadenylate-dependent ribonuclease L; and a gene within a region of linkage on chromosome 8 that encodes for macrophage scavenger receptor, have all been associated with increased risk for development of prostate cancer.[13-16] A variety of risk factors have been identified for prostate cancer, including advancing age, diet, family history and race.[13,17-19] It is known, for example, that African-American men and black African men in the UK have increased susceptibilities to the development of the disease compared with their Caucasian counterparts.[20] Furthermore, consumption of soy isoflavone-rich diets have been demonstrated to reduce the incidence of prostate cancer in native Asian men,[21,22] whereas progeny from east to west developed increasing incidences of clinical prostate cancer, thereby, linking the change to environmental factors, possibly related to the change from a soy-based diet.[23,24] Approximately 20% of all adult human cancers result from chronic inflammatory processes, presumably triggered by infectious agents or other environmental factors, and a role for inflammation has also been recently proposed in prostate carcinogenesis.[25-28] Current Concepts for Vaccine-mediated Treatment of Prostate Cancer In 1909, a role for immunologic control of cancer was proposed by Ehrlich when he predicted that the immune system repressed the growth of carcinomas that would otherwise occur with much greater frequency.[29] The possibility that the patient's own immune system might be stimulated in such a way as to effectively combat cancer has come under renewed interest recently. Although the very existence of a tumor suggests failure to generate an effective immune response, a number of studies provide evidence that vaccination is a safe and effective means to both prevent and treat some cancers. Immunotherapy is an emerging approach to treatment of prostate cancer. Two main approaches have met with at least some success: pulsing of dendritic cells with specific antigens; and vaccination of patients with allogeneic prostate cancer cells. The pulsing of dendritic cells with specific antigens is a novel approach to the treatment of prostate cancer. In general, dendritic cells are harvested from the patient, pulsed with relevant antigen(s), and returned to the patient. The APC8015 vaccine (Provenge®; Dendreon, Inc., WA, USA) is constructed of autologous dendritic cells pulsed with a fusion protein containing human prostatic acid phosphatase (PAP). Following successful Phase I and II clinical trials that demonstrated tolerance by patients,[30-34] a Phase III trial showed a median survival benefit of approximately 4.5 months in Provenge-treated patients compared with placebo-treated control patients.[35] Most recently, the US FDA has issued a Complete Response Letter regarding the biologics license application submitted by Dendreon, Inc. for Provenge (sipuleucel-T) vaccine for the treatment of asymptomatic, metastatic, androgen-independent prostate cancer.[201] The use of whole tumor cells as vaccine components allows a greatly increased antigenic menu to be presented to the immune system. Although many antigenic moieties may be unidentified, it is presumed that the rich choice of antigenic targets facilitates the likelihood of a successful immune response. Moreover, clinical models of prostate cancer immunotherapy have benefited from adenoviral vector-mediated in situ gene strategies that upregulate gene expression specifically within prostate and prostate cancer cells. Expanding and adoptively transferring prostate antigen-specific immune cells ex vivo has been important in order to circumvent immunological tolerance established at some tumor sites. In a Phase I clinical trial, Simons et al. vaccinated post-prostatectomy patients with metastatic prostate cancer with autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF),[36] a potent stimulator of systemic antitumor immunity.[37] Although some minimal side effects were observed, the authors were encouraged by an immune response characterized by enhanced delayed-type hypersensitivity (DTH) responses, in which effector cells characteristic of both Th1 and Th2 responses infiltrated into the DTH sites. Furthermore, immunoblots containing protein extracts from cultured prostate cancer cells demonstrated that some patients developed antibody responses to three extract polypeptides. However, in spite of these encouraging results, the authors concluded that clinical application of this approach was limited by the low yield of autologous cancer cells that were needed for production of the vaccine, and Phase II clinical trials were not pursued for that reason. Picking up on the potential demonstrated by autologous whole-cell vaccines, et al. investigated the utility of allogeneic whole-cell prostate cancer vaccination.[38] They reasoned that because tumor antigens are often conserved between tumors, allogeneic vaccines might stimulate cross-protective immunity.[39-41] To test this idea, monthly intradermal administrations of a vaccine composed of three irradiated allogeneic prostate cancer cell lines were given for 1 year to patients with progressive disease as defined by two consecutive increases in prostate-specific antigen (PSA). The treatment did not produce any signs of toxicity and resulted in decreased PSA velocity, as well as a cytokine response profile consistent with a Th1 immune response. In addition, median time to disease progression was 58 weeks compared with 28 weeks for historical controls. In an extension of this work, two human prostate cancer cell lines, LN-CaP and PC-3, were engineered to secrete GM-CSF and irradiated to construct the GVAX® prostate cancer vaccine (Cell Genesys, Inc., CA, USA). A Phase I clinical trial demonstrated T- and B-lymphocyte responses against autologous tumor antigens in patients vaccinated with the GVAX vaccine.[36] In a follow-up Phase I/II trial, patients demonstrated significantly reduced PSA velocity; dendritic cell and macrophage infiltrates at vaccination sites; and antibody responses to at least five antigens present in LN-CaP and PC-3 cells.[42] Furthermore, only mild side effects associated with vaccination were noted. In multicenter Phase II clinical trials involving patients with hormone-refractory prostate cancer, median survivals of 26.2 and 35.0 months in GVAX-treated patients were reported to compare favorably with a median survival of 18.9 months in patients receiving the standard of care, docetaxel plus prednisolone.[202] The GVAX prostate cancer vaccine is presently under continued evaluation in a Phase III clinical trial. The use of proapoptotic gene immunotherapy has also provided some promising results in preclinical models of prostate cancer. Transduction of neoplastic cells with 'suicide' genes allows for the ex vivo conversion of an inactive prodrug into toxic metabolites. Gene-directed enzyme prodrugs have been effective therapeutically against a variety of tumor cell types, with the directed cytotoxicity being especially practical with respect to the prostate, a dispensable organ. An important byproduct of the therapy is a localized immune response, making it a potential vaccination strategy. Several labs have recently demonstrated that proapoptotic whole tumor-cell vaccines can induce a significant (superior to traditional irradiated whole-cell vaccines) immune responses against orthotopic prostate cancer cell tumors in rodent models.[43-45] Contribution of Stroma to Tumor Growth & Progression An emerging body of evidence shows that dynamic epithelial-stromal interactions in solid tumors may select subsets of stromal cells with the ability to modulate tumor behavior, and that the local microenvironment promotes emergence of tumor-associated stromal cells with functions different from those of the normal stroma.[46-49] For example, fibroblasts derived from breast tumors stimulated morphogenesis and growth of breast preneoplastic epithelial cells, while fibroblasts derived from normal breast tissue inhibited this process.[49] Such functional changes in tumor stroma may be derived partly from the changes in secretion of growth factors and in the extracellular matrix (ECM).[50,51] The stroma consists of a mixture of cells, including fibroblasts, myofibroblasts, glial, epithelial, fat, blood, vascular, and smooth muscle cells; and the ECM, including extracellular molecules such as cytokines and growth factors (Figure 1). Of these, the predominant cell type within tumor stroma is the fibroblast. In some cancers, fibroblasts constitute a greater proportion of the overall tumor than do the neoplastic cells. Cancer-associated fibroblasts have been theorized to originate from cancer cells undergoing epithelial-to-mesenchymal transition; marrow-derived cells that have undergone migration to, and activation at, the site of the tumor; and resident fibroblasts that have undergone activation induced by neoplastic cells. In any case, cancer-associated fibroblasts are functionally and phenotypically distinct from normal fibroblasts. Fibroblasts engineered to secrete high levels of HGF or TGF-ß initiated cancer at divergent sites, including the stomach, prostate and breast in rodents.[52,53] Further, cancer-associated fibroblasts produce a number of factors that promote proliferation and progression of cancer. Among these factors are osteonectin, VEGF, and matrix metalloproteinases (MMPs).[54-56] VEGF, for example, has been implicated in a number of aspects of cancer growth, including angiogenesis, remodeling of the ECM, generation of inflammatory cytokines and hematopoietic stem cell development.[57,58] Figure 1. Tumors are complex mixtures of cell types and extracellular factors. Neoplastic cells elaborate factors that stimulate growth and angiogenesis, and inhibit apoptosis of supporting stromal tissue. In turn, the stromal tissue, which is composed of extracellular matrix and a variety of cells, including fibroblasts, vascular endothelium and smooth muscle, elaborates factors that stimulate growth of neoplastic cells, tumor vascularization and inflammation, and inhibit apoptosis. Of importance to cancer vaccination, tumor stromal cells generate an environment in which neoplastic cells are exposed to growth factors while avoiding immune recognition. This is accomplished by elaboration of cytokines that promote chronic inflammation and events leading to immune tolerance. For example, thrombospondin-1 is produced by stromal cells and leads to immune suppression via activation of TGF-ß.[59] There is also an abundance of evidence suggesting that, during tumor formation, stromal elements 'hide' or protect erroneously proliferating cells from destruction by the immune system. Via induction of local hypoxia, deregulated cytokines and a reduction in the local pH, the tumor microenvironment appears to quiet, adaptive immune responses normally generated by tumor associated antigens of malignant cells.[60,61] Tumor regression fails to occur consistently despite upregulated T-cell responses found peripherally,[62,63] substantiating the belief that trafficking of T cells directly to malignant cells is inhibited by aspects of tumor stroma. Other studies have shown that, even when local trafficking of T cells is functional, the stroma is still capable of limiting T-cell effector function.[64-66] Finally, regulatory T-cell (CD4+, CD25+) induction from within the microenvironment can reduce immune recognition of neoplastic cells and, thus, encourage tumor growth.[67-72] The Idea Behind Tissue Vaccines Beyond the neoplastic cells within a tumor, the connective tissue stroma represents an enormously unexploited reservoir of potentially powerful antigens for cancer immunotherapy. Indeed, in some carcinomas the stromal compartment may account for up to 90% of the tumor mass.[73] Tissue vaccines are constructed directly from harvested tumor material, thus, including not only cancer cells but connective tissue stroma as well (Figure 2). Figure 2. Components of cultured cell vaccines versus tissue vaccines. (A) Whole-cell tumor vaccines typically consist of one or several clonal, cultured cell lines. ( By contrast, tissue vaccines comprise a vast menu of antigenic targets, including those offered by neoplastic (cancer) cells, extracellular matrix, connective tissue cells (such as fibroblasts), inflammatory cells, and elaborated extracellular factors that promote tumor growth and metastasis. A major difficulty with cancer vaccination has been the genetic and phenotypic plasticity of many tumors. Through a number of mechanisms, cancer cells can develop means to escape immunosurveillance and destruction.[74-76] In contrast to cancer cells, however, tumor stroma cells are genetically more stable and should, therefore, represent targets that are less able to escape destruction by the immune system.[73] Furthermore, because many tumor stroma-associated antigens are upregulated or expressed only in the tumor microenvironment, they represent highly unique moieties that are unlikely to be recognized as 'self' antigens. Against this backdrop, vaccines created from harvested tissue, including stroma, create an opportunity to overcome problems associated with immunotolerance and lack of sufficient antigenic choice.[77,78] Since they are composed of material harvested directly from tumors, an additional advantage of tissue vaccines is that they include antigens expressed following in vivo growth versus the more limited antigenic profile of cultured cells. For example, cultured renal carcinoma cells showed reduced expression of a variety of genes, including some known to be tumor-associated antigens.[79] Similarly, gene expression profiling of human A549 lung adenocarcinoma cells grown in immunodeficient mice demonstrated selective induction and overexpression of genes important in tumor progression compared with cells grown in vitro.[80,81] In addition, differential expression of genes in glioma cells was associated with differences in cell migration in vivo versus in vitro.[82] The possibility that unique, important antigens can be included offers intriguing possibilities for the use of tissue vaccines versus vaccines composed of cancer cells grown or maintained in culture. Although a number of investigations have been conducted studying the use of autologous whole-cell vaccines, such as that described earlier for prostate cancer,[36] many of those efforts involved the culturing of harvested cells and thus, such vaccines do not truly constitute tissue vaccines. By contrast, few studies have examined the use of tissue vaccines for several types of cancer. Survival and freedom from tumor recurrence benefits were demonstrated for some patients vaccinated with irradiated cell suspensions from resected stage I lung tumors, although the authors concluded that the small number of autologous tumor cells that could be harvested for vaccine production meant that the probable utility of this approach would be limited.[83] A tissue lysate vaccine was used in a Phase III trial for the treatment of renal cancer when combined with nephrectomy.[84-86] In that study, the vaccine was well tolerated and 70-month progression-free survival was 72% versus 59.3% for the control group. Homogenized, formalin-fixed autologous tumor tissue was used to vaccinate patients following surgical resection of hepatocellular carcinoma.[87] Using that approach, 17 out of 24 patients developed a DTH response against vaccine components, and 1-, 2- and 3-year recurrence rates were approximately half those of nonvaccinated patients. In a limited clinical study involving two patients, treatment with alcohol was used to inactivate resected breast carcinomas prior to reimplantation at the original site.[88] Neither patient showed tumor recurrence, one at 9 years and the other at 3 years post-treatment. Laucius et al. investigated the use of an irradiated tissue vaccine adjuvanted with bacillus Calmette-Guérin (BCG) as an approach to melanoma.[89] Out of 18 patients who were vaccinated following surgery for tumor removal, four demonstrated clinical responses to vaccination. Others have further investigated this approach, although production of the vaccine used involved enzymatic digestion of the connective tissue stroma; harvested tumor tissue was treated with collagenase and DNase to produce material that was inactivated by irradiation and adjuvanted with BCG to produce a vaccine. Out of 40 patients with measurable metastases, four had complete responses and one had a partial response to vaccination; and a positive DTH response was observed in patients exhibiting tumor regression.[90] Vaccination of stage III patients who underwent surgical removal of tumor and metastases resulted in a 5-year survival rate of nearly 50%, higher than that of historical controls for patients treated only by surgery.[91,92] Hapten modification of this vaccine preparation has been shown to further improve clinical responses of patients.[93,94] In contrast to these encouraging data using a melanoma tissue vaccine, great effort was subsequently given to development of vaccines using cultured allogeneic melanoma cells. Although results were initially encouraging, two randomized adjuvant therapy trials of an allogeneic whole-cell melanoma vaccine adjuvanted with BCG were halted when it became evident that the trials were unlikely to show a significant benefit in favor of the vaccine arm.[95] This event underscores the potential efficacy of tissue vaccines versus those vaccines composed of cultured cell lines, which present a more limited scope of antigen targets to the immune system. Tumor Stromal Targets for the Antitumor Immune Response The presence of stromal components is one feature that distinguishes tissue vaccines from other whole-cell vaccines. As discussed earlier, it is evident that the stroma plays an important role in the development and progression of tumors; and, in contrast to neoplastic cells, stromal elements tend to be far more genetically stable and less apt to immune evasion. It therefore stands to reason that immune destruction of the supporting stroma should limit the ability of a tumor to advance. Several tumor stroma cell types present attractive targets for immunotherapy. Among these are tumor fibroblasts, tumor macrophages and tumor endothelial cells. Many of these cells are initially recruited by the tumor from normal surrounding tissue early in the tumorigenic process. Once recruited into the service of the advancing tumor, stromal cells are typically activated relative to their normal counterparts so that the altered stroma and ECM are conducive to tumor growth and progression. As part of this process, tumor stromal cells undergo upregulation or induction of unique antigens that may be employed to encourage immune recognition. Although data on the precise immunogenicity of individual stromal antigens are limited, the antigenic complexity offered by the stromal network is powerful.[77,78] Studies to examine the immunogenic potential of such antigens are needed to define what, if any, contribution they make to the protective immune response. A variety of molecules overexpressed by tumor stromal cells contribute, perhaps individually and certainly collectively, to the overall utility of tissue vaccines. For example, fibroblast activation protein-α is a membrane serine protease, expressed principally in fibroblasts of epithelial tumors and healing tissue, which promotes formation of tumor stroma and has been found to be a potential target for specific monoclonal antibody therapy in colon cancer.[96-100] Neoplastic cells, tumor fibroblasts, tumor endothelial cells and tumor macrophages all express MMPs, important modulators of the ECM; and MMP expression is associated with an invasive tumor phenotype and progression of the tumor.[101-103] Urokinase plasminogen activator (UPA), its associated receptor (UPAR) and inhibitor (PAI-1) are overexpressed by neoplastic cells, tumor fibroblasts, tumor endothelial cells and tumor macrophages in a variety of tumor types, including renal, breast, prostate and colorectal.[73,104-106] UPA, UPAR, and PAI-1 all play roles in cancer invasion and metastasis, and the absence of UPA and PAI-1 in genetically modified mice has been associated with impaired growth of transplanted T241 fibrosarcoma.[107] It can be noted then, that a number of tumor stroma-associated antigens are expressed exclusively in the micromilieu of the tumor and are expressed by both neoplastic and tumor stromal cells. In addition to the examples above, carbonic anhydrase IX is expressed on tumor-associated fibroblasts and on some cancer cells[108]; and survivin, a protein inhibitor of apoptosis, is overexpressed in neoplastic and in tumor epithelial cells.[109] Clearly, the tumor stroma provides an antigenically rich repertoire with which to engage the immune system. That some of the antigens are expressed by stromal components, as well as by neoplastic cells, only reinforces the closely coordinated functions of each. The neoplastic cells of the tumor are reliant upon the stromal component to modify the ECM and to support the growth and spread of the tumor. Indeed, to target the stroma is to target the entire tumor. Tissue Vaccines for Prostate Cancer While several groups have examined the use of allogeneic whole-cell vaccines or vaccines based upon pulsing of autologous dendritic cells with antigen for the treatment of prostate cancer, few have considered the potential utility of tissue vaccines for prostate cancer. In this regard, we have undertaken studies in the Lobund-Wistar (LW) rat model of prostate cancer[110] to examine the possibility that tissue vaccines are effective at both preventing and treating metastatic prostate cancer. The LW rat model of prostate adenocarcinoma was originally recognized in a colony of germfree inbred Wistar strain rats, initially in the 37th generation. Large adenocarcinomas developed in the prostatic complex spontaneously in male LW rats at approximately 24 months of age; in approximately 30% of male LW rats within 12 months following a single intravenous dose of methylnitrosourea (MNU); and in approximately 80% of male LW rats within 10 months following a single intravenous dose of MNU and subcutaneous implantation of slow-release testosterone pellets (Figure 3).[110] In addition, a transplantable cell line, PAIII, was isolated from a LW rat with spontaneous, metastasized prostate cancer and can be used to generate large, androgen-independent subcutaneous tumors following subcutaneous administration to LW rats. In all of these model systems, tumors will metastasize to the lungs via the lymphatic system. Great homology exists with the human disease in that autochthonous tumors undergo transition from an initial androgen-dependent phase to an androgen-independent phase. Moreover, prostate tumors in the LW rat express PSA, a marker sometimes associated with prostate cancer in humans.[111] Figure 3. Section of a prostate tumor from a Lobund-Wistar rat. The mass is a typical adenocarcinoma with scattered acinar structures in an abundant connective tissue stroma. Section stained with hematoxylin and eosin. Magnified x1000. The possibility that prostate cancer can be prevented by means of a prophylactic vaccine would be of tremendous value. Using tissue vaccines composed of lysates or glutaraldehyde-fixed whole-cell preparations from harvested subcutaneous PAIII tumors, we demonstrated 50 and 90% reductions, respectively, in the incidence of MNU-induced autochthonous prostate cancer in the LW rat (Figure 4).[112] When PAIII cells were coincubated with splenocytes from vaccinated LW rats prior to implantation into naive rats, 80 and 40% reductions in the incidence of subcutaneous tumors were associated with lysate and whole-cell tissue vaccination, respectively, indicating that the spleen played an important role in the observed protective immune response. We have further shown that vaccination of LW rats bearing autochthonous prostate tumors with a whole-cell tissue vaccine resulted in complete regression of the primary tumor in 21% of rats, and a 70% reduction in the incidence of rats having any evidence of pulmonary metastases. In addition, when used as an adjunct to external-beam radiation treatment of subcutaneous PAIII tumors, pretreatment with a tissue vaccine resulted in significant reduction in tumor size compared with treatment with radiation or tissue vaccine alone (Suckow MA, unpublished observations). Figure 4. Prevention of prostate cancer by tissue vaccines in Lobund-Wistar rats. Prior to tumor induction with methylnitrosourea, and monthly afterwards, rats were vaccinated with either media (as a control); a PTE of tumor material harvested from the tumors of other rats (PTE); or GFT material harvested from other rats. Compared with controls, PTE vaccination reduced the incidence of prostate cancer by 50% and GFT cell vaccination reduced the incidence by 90%. GFT: Glutaraldehyde-fixed tumor; PTE: Potassium thiocyanate extract. Ideally, a tissue vaccine would include the antigenic complexity associated with a growing tumor and be adjuvanted by the cytokine environment of such a tumor. Nonproliferating, live whole tumor-cell vaccines maintain the ability to secrete cytokines for a short period of time, yet are also processed more rapidly[113,114] and elicit a weaker immunogenic response than their live counterparts.[43,113,115-118] As discussed previously, the use of gene therapy to regulate the onset of an apoptotic death after vaccine administration approximates some aspects of a tissue vaccine. Still, even though such live, eventually apoptotic, cells exhibit efficacy, cancer cells that die a necrotic death may be better able to invoke a systemic antitumor response.[116,119-121] With this consideration, we developed a strategy that involved administration of genetically modified PAIII cells engineered to be deficient in tissue factor (TF).[122] TF is a transmembrane glycoprotein that is a critical component in the cascade initiating blood coagulation/hemostasis. Hypercoagulation has been well documented in a variety of cancers, and TF expression, specifically, has been correlated with the aggressive growth and spread of cancer.[121-128] Interference with the anti-TF pathway resulted in increased pulmonary metastasis in an experimental model of melanoma, indicating an important role for TF in the escape and migration of neoplastic cells.[129,130] Following subcutaneous administration of TF-deficient PAIII cells into naive LW rats, small tumors grow which spontaneously regress, via necrosis rather than apoptosis, within 21 days. When rats were subsequently challenged with wild-type PAIII cells, the resulting tumors were significantly smaller in vaccinated versus nonvaccinated rats and the mean number of metastatic foci in the lungs was reduced by 62.5%.[122] Xenogeneic Tissue Vaccines Clinical investigations into the use of autologous vaccines derived from harvested prostate tumor tissue led Simons et al.[36] to the conclusion that, even after expansion in culture, there was simply insufficient material for production of a vaccine quantity that would allow initial and booster doses. While the authors of that study were encouraged by the immunologic responses resulting from immunization with their preparation, which was augmented by manipulation of the autologous cells to secrete GM-CSF, future work focused on the use of cultured allogeneic prostate cancer cell lines as a source of vaccine material that could be replenished readily.[42] As a result, attention has drifted away from the substantial potential that tissue vaccines may hold for the treatment of cancer. The obvious conundrum with tissue vaccines is that, typically, the material harvested from resection or biopsy is insufficient to allow both histopathologic evaluation and production of vaccine in quantities sufficient to sustain a complete, and possibly long, course of treatment. Yet, while culture of harvested tumors might expand the quantity of raw material, by definition this weakens a main feature of the tissue vaccine: the presence of novel antigens that might be expressed in vivo but not in vitro. One possible means of overcoming the limitations to tissue vaccines posed by insufficient harvests of tumor tissue from patients, might be to use tumor tissue from other species. If homology exists between even some key antigens, such xenogeneic tissue vaccines would represent a very novel approach to cancer treatment. The idea that antigens associated with tumors from one species might stimulate protective immunity to a tumor in another species is not entirely new. For example, immunization of mice with human prostate-specific membrane antigen (PSMA) resulted in an antibody response to native mouse PSMA, suggesting that the xenogeneic antigen was sufficiently different to overcome immunologic tolerance, yet sufficiently similar to generate a cross-reactive immune response.[131] Fong et al. described the administration of mouse PAP-pulsed dendritic cells to patients with advanced prostate cancer.[132] In that study, patients developed T-cell proliferative responses to the homologous self-antigen, and some patients had stabilization of previously progressing disease. Dogs with stage II-IV melanoma were vaccinated with human tyrosinase DNA.[133] Of nine vaccinated dogs, three developed a measurable increase in postvaccine serum antibody to human tyrosinase; one of these experienced a remission with complete disappearance of radiographic evidence of pulmonary metastases; and another of the three responders had complete gross and histopathologic remission of the disease. Vaccination of stage IV colorectal cancer patients with cultured mouse B16 melanoma and lung carcinoma cells was shown to stimulate a significant increase in cell-mediated immunoreactivity as evidenced by DTH reactions, as well as by blood lymphocyte proliferation assays. Furthermore, the mean survival of vaccinated patients was 17 months versus only 7 months for nonvaccinated patients.[134] To investigate the possibility that a xenogeneic prostate tumor tissue vaccine could prevent the growth of human prostate cancer cells, we vaccinated immunocompetent BALB/c mice with a glutaraldehyde-fixed tissue vaccine prepared from PAIII prostate tumors harvested from LW rats.[135] Splenocytes harvested from vaccinated mice were incubated with PC346C human prostate cancer cells before orthotopic implantation into the prostates of syngeneic immunodeficient nude mice. After 10 weeks, we found a 70% reduction in the incidence of prostate cancer in the mice implanted with PC346C cells that had been coincubated with splenocytes from vaccinated mice, versus mice implanted with PC346C cells that had been coincubated with splenocytes from nonvaccinated control mice (Figure 5). Furthermore, cytokine profiles of cultured splenocyte supernatants suggested that the protective immunity resulted primarily from a Th1 response, with significant increases in TNF-α, IL-2 and IFN-γ, although weak serum antibody responses to a lysate of PC346 cells were detected in mice vaccinated with glutaraldehyde-fixed prostate tumor tissue cells. These data strongly suggest that xenogeneic tissue vaccines could be used to treat cancer in humans. Figure 5. Inhibition of human PC346C prostate cancer cell growth in mice by a xenogeneic tissue vaccine. Human PC346C prostate cancer cells were coincubated with splenocytes from immunocompetent BALB/c mice that had been vaccinated with GFT harvested from Lobund-Wistar rats. After incubation, the cells were orthotopically implanted into the prostates of syngeneic nude mice. After 10 weeks, prostates were evaluated for tumor growth. The graph shows that GFT vaccination stimulated immunity sufficient to reduce the incidence of prostate cancer in the mice by nearly 70% compared with nonvaccinated and media-vaccinated controls. GFT: Glutaraldehyde-fixed prostate tumor tissue. The advantages of xenogeneic tissue vaccines are twofold. First, they allow use of a replenishable source of raw material for vaccine production. The amount of vaccine available is limited only by the number of animals maintained for tumor harves. Second, and perhaps most importantly, compared with the use of autologous vaccines, xenogeneic vaccines present a mechanism by which immunotolerance can be overcome. Similar to the robust immune response associated with rejection of transplanted organs from other species, xenogeneic tissue vaccines will not be recognized as 'self' and should generate vigorous immunity to homologous antigens. Expert Commentary The standard of care for many cancers involves harsh treatments with chemotherapeutic agents, which often leave the patient clinically frail and subject to adverse sequelae. By contrast, immunotherapy offers a means to rally the body's own defenses against the cancer. While most cancer vaccines have been well tolerated by patients, they have proved to be of inconsistent clinical value. One substantial limitation of many cancer vaccines is the limited menu of antigen targets presented to the immune system. Since cancer can be a plastic, evolving tissue, the antigenic milieu is not static and vaccines composed of the broadest possible antigenic repertoires stand the best chance of success. Tissue vaccines, by their very nature, are antigenically diverse and include antigens not only from tumor neoplastic cells, but also from the supportive stromal matrix of the tumor; and they include relevant antigens that are expressed in vivo, but not in vitro. If administered as a live cell preparation that grows into a tumor, recruits a stromal matrix and then spontaneously regresses, the immune system is presented with antigens of a growing, evolving tumor. If administered as a xenogeneic preparation, the immune system is presented with antigens that may be homologous but are recognized as foreign, thus, breaking immunologic tolerance. Together, these features ascribe an antigenic richness to tissue vaccines that cannot be achieved by any other means. In some ways, tissue vaccines are not new. Early vaccines for some infectious pathogens were constructed as tissue vaccines because the pathogens could only be grown in sufficient quantities using in vivo systems. For example, formalin-fixed lung homogenates of moribund guinea pigs infected with lymphocytic choriomeningitis virus were shown to reduce mortality when given as a vaccine to naive guinea pigs subsequently challenged with lymphocytic choriomeningitis virus[136,137]; and vaccines made from formalinized tissues of either foxes or ferrets infected with distemper virus reduced mortality in naive foxes subsequently challenged with distemper virus.[138] While most cancer cells can readily be grown in culture, tumor tissue, with its complex composition of neoplastic cells and supporting stromal matrix, cannot be precisely replicated and expanded in vitro. For this reason, tumor material harvested from animal models may represent a means to rapidly produce sufficient quantities of tissue vaccine. Although the advantages of xenogeneic tissue vaccines are obvious, it would also be possible to expand, in immunodeficient rodents, tumor material harvested from patients either by resection or biopsy. In this way, then, one could generate quantities of tissue vaccine sufficient to allow treatment of patients with what would essentially be autologous tissue vaccines. In spite of significant potential advantages offered by xenogeneic tissue vaccines, several hurdles remain. First, the mechanism of action for such vaccines needs to be established. While studies in mice vaccinated with a rat-derived tissue vaccine suggest a strong role for Th1 immunity, that response needs to be characterized in greater detail and the contribution of humoral immunity to the protective response investigated further. Second, the potential for autoimmunity needs to be examined. In both rats and mice, immunization with a rat-derived tissue vaccine did not result in any notable untoward effects typical of autoimmune disease; however, studies designed to specifically examine such features need to be conducted. Finally, clinical studies must be undertaken to determine the utility of xenogeneic tissue vaccines in human patients. Although the preclinical data are compelling, translation to the clinic will require careful analysis in human clinical trials. Five-year View We believe that the significant need for novel approaches to cancer may benefit from the further development of tissue vaccines. Potentially, use of tissue vaccines or other types of cancer vaccines might be used as part of a standard immunologic prophylaxis program against those cancers for which an individual patient may be at greatest risk. That tissue vaccines can be used as autologous preparations, xenogeneic preparations, or as vaccines that immunize via induction of a small, spontaneously regressing tumor, supports the tremendous potential of this approach to cancer. With proven efficacy in preclinical models, including one which utilized a human cancer cell line, further research with tissue vaccines should involve refinement by added adjuvants and use of novel vaccine-delivery systems; and, ultimately, human clinical testing. Determining optimal administration conditions will be necessary to avoid encouragement of host immune regulatory mechanisms and an immunosuppressive tumor environment.[139] While such testing will need to establish the safety of xenogeneic tissue vaccines and the relative contribution of antitumor stroma immunity, our demonstration of efficacy suggests that a relatively unlimited supply of vaccine material obtained from tumors of other species could overcome the hurdle of insufficient autologous material described by Simons et al..[36] The possibility that xenogeneic material could be used to generate vaccines makes Simons' encouraging results a more translational possibility. References A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. Cancer statistics, 2007. CA Cancer J. Clin. 57(1), 43-66 (2007). Greene KL, Cowan JE, berg MR et al. Who is the average patient presenting with prostate cancer? Urology 66(5 Suppl.), 76-82 (2005). Adolfsson J, Tribukait B, Levitt S. The 20-yr outcome in patients with well- or moderately differentiated clinically localized prostate cancer diagnosed in the pre-PSA era: the prognostic value of tumour ploidy and comorbidity. Eur. Urol. 52(4), 1028-1035 (2007). Boyd NF, Clemens JD, Feinstein AR. Pretherpeutic morbidity in the prognostic staging of acute leukemia. Arch. Intern. Med. 139(3), 324-328 (1979). Satariano WA, Ragland KE, Van Den Eeden SK. Cause of death in men diagnosed with prostate carcinoma. Cancer 83(6), 1180-1188 (1998). Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J. Chronic Dis. 40(5), 373-383 (1987). Litwin MS, Greenfield S, Elkin EP, Lubeck DP, Broering JM, Kaplan SH. Assessment of prognosis with the total illness burden index for prostate cancer: aiding clinicians in treatment choice. Cancer 109(9), 1777-1783 (2007). Albertsen PC, Fryback DG, Storer BE, Kolon TF, Fine J. The impact of co-morbidity on life expectancy among men with localized prostate cancer. J. Urol. 156(1), 127-132 (1996). Stier DM, Greenfield S, Lubeck DP et al. Quantifying comorbidity in a disease-specific cohort: adaptation of the total illness burden index to prostate cancer. Urology 54(3), 424-429 (1999). Arredondo SA, Elkin EP, Marr PL et al. Impact of comorbidity on health-related quality of life in men undergoing radical prostatectomy: data from CaPSURE. Urology 67(3), 559-565 (2006). Gonzalgo M, Isaacs WB. Molecular pathways to prostate cancer. J. Urol. 170(6 Pt 1), 2444-2452 (2003). Shand R, Gelmann EP. Molecular biology of prostate-cancer pathogenesis. Curr. Opin. Urol. 16(3), 123-131 (2006). Cussenot O, Valeri A, Berthon P, Fournier G, Mangin P. Hereditary prostate cancer and other genetic predispositions to prostate cancer. Int. J. Urol. 60(Suppl. 2), 30-34 (1998). Verhage BA, Kiemeney LA. Inherited predisposition to prostate cancer. Eur. J. Epidemiol. 18(11), 1027-1036 (2003). Gronberg H, J, Emanuelsson M et al. In Swedish families with hereditary prostate cancer, linkage to the HPC1 locus on chromosome 1q24-25 is restricted to families with early-onset prostate cancer. Am. J. Hum. Genet. 65(1), 134-140 (1999). Simard J, Dumont M, Labuda D et al.Prostate cancer susceptibility genes: lessons learned and challenges posed. Endocr. Relat. Cancer 10(2), 225-259 (2003). Nomura AMY, Lee J, Stemmermann GN, Combs GF. Serum selenium and subsequent risk of prostate cancer. Cancer Epidemiol. Biomarkers Prev. 9(9), 883-887 (2000). Hursting SD, Thornquist M, MM. Types of dietary fat and the incidence of cancer at five sites. Prev. Med. 19(3), 242-253 (1990). Matchariyakul C, Kochakarn W, Chaimuangraj S, Leeanupunth C, Lertsithichai P. A risk index for prostate cancer. Int. J. Urol. 11(5), 310-315 (2004). Ben-Shlomo Y, S, Ibrahim F et al. The risk of prostate cancer amongst black men in the United Kingdom: the PROCESS cohort study. Eur. Urol. DOI:10.1016/j.eururo.2007.02.047 (2007) (Epub ahead of print). Haas GP, Sakr WA. Epidemiology of prostate cancer. CA Cancer J. Clin. 47(5), 273-287 (1997). IJ, Banerjee M, Sakr W et al. Should African American men be treated for prostate cancer at an earlier age than white men? Cancer 85(2), 472-477 (1999). Haenszel W, Kurihara M. Studies of Japanese migrants. I. Mortality from cancer and other diseases among Japanese in the United States. J. Natl Cancer Inst. 40(1), 43-68 (1968). Kolonel LN. Variability in diets and its relation to risk in ethnic and migrant groups. Basic Life Sci. 43(1), 129-135 (1988). Ames BN, Gold LS, Willett WC. The causes and prevention of cancer. Proc. Natl Acad. Sci. USA 92(12), 5258-5265 (1995). Coussens LM, Werb Z. Inflammation and cancer. Nature 420(6917), 860-867 (2002). Rollins BJ. Inflammatory chemokines in cancer growth and progression. Eur. J. Cancer 42(6), 760-767 (2006). De Marzo A, Platz EA, Sutcliffe S et al. Inflammation in prostate carcinogenesis. Nat. Rev. Cancer 7(4) 256-269 (2007). Ehrlich P. Über den jetzigen Stand der Karzinomforschung. Ned Tijdschr Geneeskd 5(Pt 1), 273-290 (1909). Beinart G, Rini B, Weinberg VW, Small EJ. APC8015 (Provenge®) in patients with androgen-dependent biochemically-relapsed prostate cancer. Clin. Prostate Cancer 4(1), 55-60 (2005). Rini BI, Weinberg V, Fong L, Conry S, Hershberg RM, Small EJ. Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (provenge) plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy. Cancer 107(1), 67-74 (2006). Small EJ, Fratesi P, Reese DM et al. Immunotherapy of hormone-refractory prostate cancer with antigen-loaded dendritic cells. J. Clin. Oncol. 18(23), 3894-3903 (2000). Burch PA, Breen JK, Buckner JC et al. Priming tissue specific cellular immunity in a Phase I trial of autologous dendritic cells for prostate cancer. Clin. Cancer Res. 6(6), 2175-2182 (2000). Burch PA, Croghan GA, Gastineau DA et al. Immunotherapy (APC8015, Provenge) targeting prostatic acid phosphatase can induce durable remission of metastatic androgen-independent prostate cancer: a Phase 2 trial. Prostate 60(3), 197-204 (2004). Small EJ, Schellhammer PF, Higano CS et al. Placebo-controlled Phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J. Clin. Oncol. 24(19), 3089-3094 (2006). Simons JW, Mikhak B, Chang JF et al. Induction of immunity to prostate cancer antigens: results of a clinical trial of vaccination with irradiated autologous prostate tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor using ex vivo gene transfer. Cancer Res. 59(20), 5160-5168 (1999). Dranoff G, Jaffee E, Lazenby A et al. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc. Natl Acad. Sci. USA 90(8), 3539-3543 (1993). A, Ball G, Quatan N et al. Delayed disease progression after allogeneic cell vaccination in hormone-resistant prostate cancer and correlation with immunologic variables. Clin. Cancer Res. 11(12), 4469-4478 (2005). Cornet S, Miconnet I, Menez J, Lemonnier F, Kosmatopoulos K. Optimal organization of a polypeptide-based candidate cancer vaccine antigen composed of cryptic tumor peptides with enhanced immunogenicity. Vaccine 24(12), 2102-2109 (2006). Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity 10(6), 673-679 (1999). Scardino A, Gross DA, Alves P et al. HER-2/neu and hTERT cryptic epitopes as novel targets for broad spectrum tumor immunotherapy. J. Immunol. 168(11), 5900-5906 (2002). Simons JW, Carducci MA, Mikhak B et al. Phase I/II trial of an allogeneic cellular immunotherapy in hormone-naïve prostate cancer. Clin. Cancer Res. 12(11), 3394-3401 (2006). Pandha HS, Cook D, Greenhalgh R, Dalgleish A. Immunotherapy of murine prostate cancer using whole tumor cells killed by ex vivo cytosine deaminase/ 5-fluorocytosine suicide-gene therapy. BJU Int. 95(9), 1336-1342 (2005). Dzojic H, Loskog A, Totterman TH, Essand M. Adenovirus-mediated CD40 ligand therapy induces tumor cell apoptosis and systemic immunity in the TRAMP-C2 mouse prostate cancer model. Prostate 66(8), 831-838 (2006). Hall SJ, Sanford MA, Atkinson G, Chen SH. Induction of potent antitumor natural killer cell activity by herpes simplex virus-thymidine kinase and gancyclovir therapy in an orthotopic mouse model of prostate cancer. Cancer Res. 58(15), 3221-3225 (1998). Fukino K, Shen L, Matsumoto S, on CD, Mutter GL, Eng C. Combined total genome loss of heterozygosity scan of breast cancer stroma and epithelium reveals multiplicity of stroma targets. Cancer Res. 64(20), 7231-7236 (2004). Bissell MJ, Barcellos-Hoff MH. The influence of extracellular matrix on gene expression: is structure the message? J. Cell Sci. Suppl. 8, 327-343 (1987). Matrisian LM, Cunha GR, Mohla S. Epithelial-stromal interactions and tumor progression: meeting summary and further directions. Cancer Res. 61(9), 3844-3846 (2001). Shekhar MP, Werdell J, Santner SJ, ey RJ, Tait L. Breast stroma plays a dominant regulatory role in breast epithelial growth and differentiation: implications for tumor development and progression. Cancer Res. 61(4), 1320-1326 (2001). Schor SL, Schor AM. Phenotypic and genetic alterations in mammary stroma: implications for tumour progression. Breast Cancer Res. 3(6), 373-379 (2001). Haslam SZ, Woodward TL. Host microenvironment in breast cancer development: epithelial-cell-stromall-cell interactions and steroid hormone action in normal and cancerous mammary gland. Breast Cancer Res. 5(4), 208-215 (2003). Bhowmick NA, Chytil A, Plieth D et al. TGF-ß signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 303(5659), 848-851 (2004). Kuperwasser C, Chavarria T, Wu M et al. Reconstruction of functionally normal and malignant human breast tissues in mice. Proc. Natl Acad. Sci. USA 101(14), 4966-4971 (2004). Sangaletti S, Stoppacciaro A, Guiducci C, Torrisi MR, Colombo MP. Leukocyte rather than tumor-produced SPARC determines stroma and collagen type IV deposition in mammary carcinoma. J. Exp. Med. 198(10), 1475-1485 (2003). Okuyama H, Krishnamachary B, Zhou YF, Nagasawa H, Bosch-Marce M, Semenza GL. Expression of vascular endothelial growth factor receptor 1 in bone marrow derived mesenchymal cells is dependent on hypoxia inducible factor 1. J. Biol. Chem. 281(22), 15554-15563 (2006). Pei D. Matrix metalloproteinases target protease-activated receptors on the tumor cell surface. Cancer Cells 7(3), 207-208 (2005). Kaplan RN, Riba RD, Zacharoulis S et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438(7069), 820-827 (2005). Li H, Fan X, Houghton J. Tumor microenvironment: the role of the tumor stroma in cancer. J. Cell. Biochem. 101 (4), 805-815 (2007). Silze T, Randolph GJ, Kreutz M, Kunz-Schughart LA. The fibroblast: sentinel cell and local immune modulator in tumor tissue. Int. J. Cancer 108(2), 173-180 (2004). Balkwill F, KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 7(3), 211-217 (2005). Henning T, Kraus M, Brischwein M, Otto AM, Wolf B. Relevance of tumor microenvironment for progression, therapy and drug development. Anticancer Drugs 15(1), 7-14 (2004). Singh S, Ross SR, Acena M, Rowley DA, Schreiber H. Stroma is critical for preventing or permitting immunological destruction of antigenic cancer cells. J. Exp. Med. 175(1), 139-146 (1992). Gajewski TF, Meng Y, Blank C et al. Immune resistance orchestrated by the tumor microenvironment. Immunol. Rev. 213, 131-145 (2006). Perdrizet GA, Ross SR, Stauss HJ, Singh S, Koeppen H, Schreiber H. Animals bearing malignant grafts reject normal grafts that express through gene transfer the same antigen. J. Exp. Med. 171(4), 1205-1220 (1990). Frey AB, Monu N. Effector-phase tolerance: another mechanism of how cancer escapes antitumor immune response. J. Leukoc. Biol. 79(4), 652-662 (2006). Ganss R, Arnold B, Hammerling GJ. Mini-review: overcoming tumor-intrinsic resistance to immune effector function. Eur. J. Immunol. 34(10), 2635-2641 (2004). Woo EY, Yeh H, Chu CS et al. Cutting edge: regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. J. Immunol. 168(9), 4272-4276 (2002). Woo EY, Chu CS, Goletz TJ et al. Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res. 61(12), 4766-4772 (2001). Sasada T, Kimura M, Yoshida Y, Kanai M, Takabayashi A. CD4+CD25+ regulatory T cells in patients with gastrointestinal malignancies: possible involvement of regulatory T cells in disease progression. Cancer 98(5), 1089-1099 (2003). Wang HY, Lee DA, Peng G et al. Tumor-specific human CD4+ regulatory T cells and their ligands: implications for immunotherapy. Immunity 20(1), 107-118 (2004). Curiel TJ, Coukos G, Zou L et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 10(9), 942-949 (2004). Wang Y, Zhou L, Wang HY, Xiao JX, Si LS, Wang YL. The ex vivo microenvironments in MLTC of poorly immunogenic tumor cells facilitate polarization of CD4+CD25+ regulatory T cells. Cell Mol. Immunol. 3(2), 123-129 (2006). Hofmeister V, Vetter C, Schrama D, Bröcker E-B, Becker JC. Tumor stroma-associated antigens for anti-cancer immunotherapy. Cancer Immunol. Immunother. 55(5), 481-494 (2006). Ahmad M, Rees RC, Ali SA. Escape from immunotherapy: possible mechanisms that influence tumor regression/progression. Cancer Immunol. Immunother. 53(10), 844-854 (2004). Pawelec G. Tumour escape: antitumour effectors too much of a good thing? Cancer Immunol. Immunother. 53(3), 262-274 (2004). thor Straten P, Becker JC, Guldberg P, Zeuthen J. In situ T cells in melanoma. Cancer Immunol. Immunother. 48(7), 386-395 (1999). Kammertoens T, Schuler T, Blankenstein T. Immunotherapy: target the stroma to hit the tumor. Trends Mol. Med. 11(5), 225-231 (2005). Hofmeister V, Schrama D, Becker JC. Anti-cancer therapies targeting tumor stroma. Cancer Immunol. Immunother. 57(1), 1-17 (2008). Moschella F, Cantanzaro RP, Bisikirska B et al. Shifting gene expression profiles during ex vivo culture of renal tumor cells: implications for cancer immunotherapy. Oncol. Res. 14(3), 133-145 (2003). Creighton CJ, Bromberg-White JL, Misek DE et al. Analysis of tumor-host interactions by gene expression profiling of lung adencarcinoma xenografts identifies genes involved in tumor formation. Mol. Cancer Res. 3(3), 119-129 (2005). Creighton C, Kuick R, Misek DE et al. Profiling of pathway-specific changes in gene expression following growth of human cancer cell lines transplanted into mice. Genome Biol. 4(7), R46 (2003). Tatenhorst L, Püttmann S, Senner V, us W. Genes associated with fast glioma cell migration in vitro and in vivo. Brain Pathol. 15(1), 46-54 (2005). Stack BHR, McSwan N, Stirling JM et al. Autologous x-irradiated tumour cells and percutaneous BCG in operable lung cancer. Thorax 37(8), 588-593 (1982). Jocham D, Richter A, Hoffmann L et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: Phase III, randomized controlled trial. Lancet 363(9409), 594-599 (2004). Repmann R, Goldschmidt AJ, Richter A. Adjuvant therapy of renal cell carcinoma patients with an autologous tumor cell lysate vaccine: a 5-year follow-up analysis. Anticancer Res. 23(2A), 969-974 (2003). Repmann R, Wagner S, Richter A. Adjuvant therapy of renal cell carcinoma with active-specific-immunotherapy (ASI) using autologous tumor vaccine. Anticancer Res. 17(4B), 2879-2882 (1997). Peng B-G, Liang L-J, He Q et al. Tumor vaccine against recurrence of hepatocellular carcinoma. World J. Gastroenterol. 11(5), 700-704 (2005). Kobayashi T. Autologous vaccine from isopropyl-alcohol denatured breast cancer tissue prevented cancer recurrence without chemotherapy or radiotherapy. J. Integr. Med. 8(1), 97-101 (2004). Laucius JF, Bodurtha AJ, Mastrangelo MJ, Bellet RE. A Phase II study of autologous irradiated tumor cells plus BCG in patients with metastatic malignant melanoma. Cancer 40(5), 2091-2093 (1977). Berd D, Maguire HC Jr, McCue P, Mastrangelo MJ. Treatment of metastatic melanoma with an autologous tumor-cell vaccine: clinical and immunologic results in 64 patients. J. Clin. Oncol. 8(11), 1858-1867 (1990). Baars A, Claessen AME, van den Eertwegh AJM et al. Skin tests predict survival after autologous tumor cell vaccination in metastatic melanoma: experience in 81 patients. Ann. Oncol. 11(8), 965-970 (2000). Essner R, Lee JH, Wanek LA, Itakura H, Morton DL. Contemporary surgical treatment of advanced-stage melanoma. Arch. Surg. 139(9), 961-966 (2004). Berd D, Maguire HC Jr, Schuchter LM et al. Autologous hapten-modified melanoma vaccine as postsurgical adjuvant treatment after resection of nodal metastases. J. Clin. Oncol. 15(6), 2359-2370 (1997). Berd D, Kairys J, Dunton C, Mastrangelo MJ, Sato T, Maguire HC Jr. Autologous, hapten-modified vaccine as a treatment for human cancers. Semin. Oncol. 25(6), 646-653 (1998). Sondak VK, Sabel MS, Mulé JJ. Allogeneic and autologous melanoma vaccines: where have we been and where are we going? Clin. Cancer Res. 12(7 Suppl.), 2337s-2341s (2006). Garin-Chesa P, Old LJ, Rettig WJ. Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers. Proc. Natl Acad. Sci. USA 87(18), 7235-7239 (1990). Niedermeyer J, Scanlan MJ, Garin-Chesa P et al. Mouse fibroblast activation protein: molecular cloning, alternative splicing and expression in the reactive stroma of epithelial cancers. Int. J. Cancer 71(3), 383-389 (1997). Park JE, Lenter MC, Zimmermann RN, Garin-Chesa P, Old LJ, Rettig WJ. Fibroblast activation protein, a dual specificity serine protease expressed in reactive human tumor stromal fibroblasts. J. Biol. Chem. 274(51), 36505-36512 (1999). Tahtis K, Lee F-T, Wheatley JM et al. Expression and targeting of human fibroblast activation protein in a human skin/severe combined immunodeficient mouse breast cancer xenograft model. Mol. Cancer Ther. 2(8), 729-737 (2003). Welt S, Divgi CR, AM et al. Antibody targeting in metastatic colon cancer: a Phase I study of monoclonal antibody F19 against a cell-surface protein of reactive tumor stromal fibroblasts. J. Clin. Oncol. 12(6), 1193-1203 (1994). Hofmann UB, Eggert AA, Blass K, Brocker EB, Becker JC. Expression of matrix metalloproteinases in the microenvironment of spontaneous and experimental melanoma metastases reflects the requirements for tumor formation. Cancer Res. 63(23), 8221-8225 (2003). Hofmann UB, Westphal JR, Van Muijen GN, Ruiter DJ. Matrix metalloproteinases in human melanoma. J. Invest. Dermatol. 115(3), 337-344 (2000). Ntayi C, Hornebeck W, Bernard P. Influence of cultured dermal fibroblasts on human melanoma cell proliferation, matrix metalloproteinase-2 (MMP-2) expression and invasion in vitro. Arch. Dermatol. Res. 295(6), 236-241 (2003). Fischer WH, thor Straten P, Terheyden P, Becker JC. Function and dysfunction of CD4(+) T cells in the immune response to melanoma. Cancer Immunol. Immunother. 48(7), 363-370 (1999). Usher PA, Thomsen OF, Iversen P et al. Expression of urokinase plasminogen activator, its receptor and type-1 inhibitor in malignant and benign prostate tissue. Int. J. Cancer 113(6), 870-880 (2005). Xu Y, Hagege J. Doublet JD et al. Endothelial and macrophage upregulation of urokinase receptor expression in human renal cell carcinoma. Hum. Pathol. 28(2), 206-213 (1997). Gutierrez LS, Schulman A, Brito- T, Noria F, Ploplis VA, Castellino FJ. Tumor development is retarded in mice lacking the gene for urokinase-type plasminogen activator or its inhibitor, plasminogen activator inhibitor-1. Cancer Res. 60(20), 5839-5847 (2000). Ivanov S, Liao SY, Ivanova A et al. Expression of hypoxia-inducible cell-surface transmembrane carbonic anhydrases in human cancer. Am. J. Pathol. 158(3), 905-919 (2001). Gianani R, Jarboe E, Orlicky D et al. Expression of surviving in normal, hyperplastic, and neoplastic colonic mucosa. Hum. Pathol. 32(1), 119-125 (2001). Pollard M, Suckow MA. Hormone-refractory prostate cancer in the Lobund-Wistar rat. Exp. Biol. Med. (Maywood) 230(8), 520-526 (2005). Pham H, Vang K, Ziboh VA. Dietary γ-linolenate attenuates tumor growth in a rodent model of prostatic adenocarcinoma via suppression of elevated generation of PGE2 and 5S-HETE. Prostaglandins Leukot. Essent. Fatty Acids 74(4), 271-282 (2006). Suckow MA, Wolter WR, Pollard M. Prevention of de novo prostate cancer by immunization with tumor-derived vaccines. Cancer Immunol. Immunother. 54(6), 571-576 (2005). Tai KF, Chen DS, Hwang LF. Curative potential of GM-CSF-secreting tumor cell vaccines on established orthotopic liver tumors: mechanisms for the superior antitumor activity of live tumor cell vaccines. J. Biomed. Sci. 11(2), 228-238 (2004). Labarthe MC, Halanek N, Birchall L et al. The biological effects of syngeneic and allogeneic cytokine-expressing prophylactic whole cell vaccines and the influence of irradiation in a murine melanoma model. Cancer Immunol. Immunother. 55(3), 277-288 (2005). Vile RG, Castleden S, Marshall J, Camplejohn R, Upton C, Chong H. Generation of an anti-tumour immune response in a non-immunogenic tumour: HSVtk killing in vivo stimulates a mononuclear cell infiltrate and a Th1-like profile of intratumoural cytokine expression. Int. J. Cancer 71(2), 267-274 (1997). Melcher A, Todryk S, Hardwick N, Ford M, son M, Vile RG. Tumor immunogenicity is determined by the mechanisms of cell death via induction of heat shock protein expression. Nat. Med. 4(5), 581-587 (1998). Todryk S, Melcher A, Bottley G, Gough M, Vile R. Cell death associated with genetic prodrug activation therapy of colorectal cancer. Cancer Lett. 174(1), 25-33 (2001). Pandha H, Eaton J, Greenhalgh R, Soars D, Dalgleish A. Immunotherapy of murine prostate cancer using whole tumor cells killed ex vivo by herpes simplex viral thymidine kinase/gancyclovir suicide gene therapy. Cancer Gene Ther. 12(6), 572-578 (2005). Basu S, Binder RJ, Suto R, KM, Srivastava PK. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-κB pathway. Int. Immunol. 12(11), 1539-1546 (2000). Gough MJ, Melcher AA, Ahmed A et al. Macrophages orchestrate the immune response to tumor cell death. Cancer Res. 61(19), 7240-7247 (2001). Proskuryakov SY, Gabai VL, Konoplyannikov AG, Zamulaeva IA, Kolesnikova AI. Immunology of apoptosis and necrosis. Biochemistry Mosc. 70(12), 1310-1320 (2005). Heinrich JE, Pollard M, Wolter WR et al. Vaccination against prostate cancer using a live tissue factor deficient cell line in Lobund-Wistar rats. Cancer Immunol. Immunother. 56(5), 725-730 (2007). Callander NS, Varki N, Rao LV. Immunohistochemical identification of tissue factor in solid tumors. Cancer 70(5), 1194-1201 (1992). Abdulkadir SA, Carvalhal GF, Kaleem Z et al. Tissue factor expression and angiogenesis in human prostate carcinoma. Hum. Pathol. 31(4), 443-447 (2000). Poon RT, Lau CP, Ho JW, Yu WC, Fan ST, Wong J. Tissue factor expression correlates with tumor angiogenesis and invasiveness in human hepatocellular carcinoma. Clin. Cancer Res. 9(14), 5339-5345 (2003). Kaido T, Oe H, Yoshikawa A, Mori A, Arii S, Imamura M. Tissue factor is a useful prognostic factor of recurrence in hepatocellular carcinoma in 5-year survivors. Hepatogastroenterology 52(65), 1383-1387 (2005). Nitori N, Ino Y, Nakanishi Y et al. Prognostic significance of tissue factor in pancreatic ductal adenocarcinoma. Clin. Cancer Res. 11(7), 2531-2539 (2005). Chand HS, Ness SA, Kisiel W. Identification of a novel human tissue factor splice variant that is upregulated in tumor cells. Int. J. Cancer 118(7), 1713-1720 (2006). Mueller BM, Reisfeld RA, Edgington TS, Ruf W. Expression of tissue factor by melanoma cells promotes efficient hematogenous metastasis. Proc. Natl Acad. Sci. USA 89(24), 11832-11836 (1992). Amirkhosravi A, Meyer T, Chang JY et al. Tissue factor pathway inhibitor reduces experimental lung metastasis of B16 melanoma. Thromb. Haemost. 87(6), 930-936 (2002). Gregor PD, Wolchok JD, Turaga V et al. Induction of autoantibodies to syngeneic prostate-specific membrane antigens by xenogeneic vaccination. Int. J. Cancer 116(3), 415-421 (2005). Fong L, Brockstedt D, Benike C et al. Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J. Immunol. 167(12), 7150-7156 (2001). Liao JCF, Gregor P, Wolchok JD et al. Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma. Cancer Immun. 6, 8 (2006). Seledtsov VI, Niza NA, Felde MA et al. Xenovaccinotherapy for colorectal cancer. Biomed. Pharmacother. 61(2-3), 125-130 (2007). Suckow MA, Rosen ED, Wolter WR, Sailes V, R, Tenniswood M. Prevention of human PC-346C prostate cancer growth in mice by a xenogeneic tissue vaccine. Cancer Immunol. Immunother. 56(8), 1275-1283 (2007). Traub E. Immunization of guinea pigs with a modified strain of lymphocytic choriomeningitis virus. J. Exp. Med. 66(3), 317-324 (1937). Traub E. Immunization of guinea pigs against lymphocytic choriomeningitis with formolized tissue vaccines. J. Exp. Med. 68(1), 95-110 (1938). Heath LM. Distemper studies in foxes. III. The cross immunizing properties of homologous and heterologous tissue vaccines against experimental distemper of foxes and ferrets. Can. J. Comp. Med. 7(9), 266-272 (1943). Sakaguchi S. Naturally arising CD4+ regulatory Tcells for immunologic self-tolerance and negative control of immune responses. Annu. Rev. Immunol. 22, 531-562 (2004). Sidebar: Key Issues Most cancer vaccines are based on single proteins or cultured cell lines. Immunotolerance is an enormous hurdle to successful application of cancer vaccines. Tissue vaccines are composed of material harvested directly from tumors. Tissue vaccines include antigens associated with neoplastic cells and with the tumor stroma. Furthermore, they include antigens that may be expressed only following in vivo growth versus growth in culture. Attempts at autologous cancer vaccines have been limited by finite, typically insufficient amounts of raw material for vaccine production. By contrast, xenogeneic tissue vaccines offer a means to generate sufficient quantities of vaccine raw material, as well as the potential to overcome immunotolerance. Cancer cells engineered to generate spontaneously regressing tumors following implantation offer a means to vaccinate the animal using antigens associated with a growing tumor. In animal models of prostate cancer, tissue vaccines reduced the incidence by 90% when used prophylactically; resulted in complete tumor regression in 20% and a 70% reduction in the incidence of metastasis in tumor-bearing animals; and stimulated immunity associated with a 70% reduction against tumor formation associated with a human prostate cancer cell line. Clinical studies are needed to assess the utility of tissue vaccines in patients. Acknowledgements The authors thank Schroeder for technical assistance in work relevant to this manuscript and for the illustrations in this manuscript. Reprint Address Mark A Suckow Associate Research Professor, Biological Sciences, University of Notre Dame, Freimann Life Science Center, Notre Dame, IN. suckow.1@... . Mark A Suckow,1 Heinrich,2 Elliot D Rosen3 1Biological Sciences, University of Notre Dame, Freimann Life Science Center, Notre Dame, IN 2University of Notre Dame, Department of Biological Sciences, Notre Dame, IN 3Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN The Great HPV Vaccine Hoax Exposed Great Newstarget.com article!! I would cut and paste the whole thing here but it is 7 pages. This link takes you to page 1 then you can click NEXT at the bottom of each page to read the whole article. http://www.newstarg et.com/Report_ HPV_Vaccine_ 1.html Diane Quote Link to comment Share on other sites More sharing options...
Recommended Posts
Join the conversation
You are posting as a guest. If you have an account, sign in now to post with your account.
Note: Your post will require moderator approval before it will be visible.