p53 tumor suppressor gene therapy for cancer · 2017-10-19 · restoring p53 function can induce...

9
p53 Tumor Suppressor Gene Therapy for Cancer Published on Patient Care Online (http://www.patientcareonline.com) p53 Tumor Suppressor Gene Therapy for Cancer Review Article [1] | October 01, 1999 By Jack A. Roth, MD, FACS [2], Stephen G. Swisher, MD [3], and Raymond E. Meyn, PhD [4] Gene therapy has the potential to provide cancer treatments based on novel mechanisms of action with potentially low toxicities. This therapy may provide more effective control of locoregional recurrence in diseases like non–small-cell lung cancer (NSCLC) as well as systemic control of micrometastases. Despite current limitations, retroviral and adenoviral vectors can, in certain circumstances, provide an effective means of delivering therapeutic genes to tumor cells. Although multiple genes are involved in carcinogenesis, mutations of the p53 gene are the most frequent abnormality identified in human tumors. Preclinical studies both in vitro and in vivo have shown that restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like cisplatin (Platinol) and ionizing radiation work synergistically to induce apoptosis in cancer cells. Phase I clinical trials now show that p53 gene replacement therapy using both retroviral and adenoviral vectors is feasible and safe. In addition, p53 gene replacement therapy induces tumor regression in patients with advanced NSCLC and in those with recurrent head and neck cancer. This article describes various gene therapy strategies under investigation, reviews preclinical data that provide a rationale for the gene replacement approach, and discusses the clinical trial data available to date. [ ONCOLOGY 13(Suppl 5):148-154, 1999] Introduction The concept of gene therapy to treat human disease originally developed as a potential treatment for inherited monogenic disorders.[1,2] In theory, a disease caused by the absence or mutation of a single gene, such as cystic fibrosis or Gauchers disease, could be treated and potentially cured by inserting a normal copy of the mutant or deleted gene into a renewable population of host cells, such as bone marrow stem cells. While conceptually simple, this strategy of gene replacement therapy is proving to have practical complexities that make its clinical implementation more difficult than had been anticipated. Currently available vectors have been unable to sustain high enough levels of gene expression over long enough periods of time. A major focus in gene therapy has been the treatment of cancer. Approaches have included transferring cytokine genes to stimulate an antitumor immune response, delivering genes that express prodrugs to tumors, and transferring genes to protect stem cells during high-dose chemotherapy. This review will focus on restoring the function of tumor suppressor genes. In the context of cancer, transient gene expression that triggers cancer cell death may be enough to mediate a therapeutic effect. Expression of the transgene in only a fraction of tumor cells also may not be limiting because these cells may alter the growth of adjacent cells.[3] Genetic Basis of Carcinogenesis The gene families implicated in carcinogenesis include dominant oncogenes like ras and tumor suppressor genes like p53.[4-6] Proto-oncogenes (normal counterparts of oncogenes) ordinarily participate in such functions as signal transduction (relaying information from the outer cell membrane to the nucleus) and gene transcription. An abnormality (point mutation, amplification, translocation, or rearrangement) in only one of the two alleles of a proto-oncogene is sufficient to convert it into an active oncogene, that is, to dysregulate its function and lead to malignant transformation. In contrast, a single normal tumor suppressor gene in a cell is typically sufficient to perform the normal function of the gene, but loss of function of both alleles, by mutation or deletion or a combination thereof, leads to dysregulation of cellular growth. In addition, some tumor suppressor gene mutationsfor example, some p53 mutationsact in a dominant negative manner, that is, a mutation in one allele can lead to production of a mutant p53 protein that binds to, and thereby inactivates, the structurally normal protein encoded by the opposite allele. Viral proteins also may bind and functionally inactivate p53.[7] Page 1 of 9

Upload: others

Post on 16-Mar-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

p53 Tumor Suppressor Gene Therapy for CancerReview Article [1] | October 01, 1999By Jack A. Roth, MD, FACS [2], Stephen G. Swisher, MD [3], and Raymond E. Meyn, PhD [4]

Gene therapy has the potential to provide cancer treatments based on novel mechanisms of actionwith potentially low toxicities. This therapy may provide more effective control of locoregionalrecurrence in diseases like non–small-cell lung cancer (NSCLC) as well as systemic control ofmicrometastases. Despite current limitations, retroviral and adenoviral vectors can, in certaincircumstances, provide an effective means of delivering therapeutic genes to tumor cells. Althoughmultiple genes are involved in carcinogenesis, mutations of the p53 gene are the most frequentabnormality identified in human tumors. Preclinical studies both in vitro and in vivo have shown thatrestoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression andDNA-damaging agents like cisplatin (Platinol) and ionizing radiation work synergistically to induceapoptosis in cancer cells. Phase I clinical trials now show that p53 gene replacement therapy usingboth retroviral and adenoviral vectors is feasible and safe. In addition, p53 gene replacementtherapy induces tumor regression in patients with advanced NSCLC and in those with recurrent headand neck cancer. This article describes various gene therapy strategies under investigation, reviewspreclinical data that provide a rationale for the gene replacement approach, and discusses theclinical trial data available to date. [ ONCOLOGY 13(Suppl 5):148-154, 1999]

Introduction

The concept of gene therapy to treat human disease originally developed as a potential treatment forinherited monogenic disorders.[1,2] In theory, a disease caused by the absence or mutation of asingle gene, such as cystic fibrosis or Gaucher�s disease, could be treated and potentially cured byinserting a normal copy of the mutant or deleted gene into a renewable population of host cells, suchas bone marrow stem cells. While conceptually simple, this strategy of gene replacement therapy isproving to have practical complexities that make its clinical implementation more difficult than hadbeen anticipated. Currently available vectors have been unable to sustain high enough levels ofgene expression over long enough periods of time.

A major focus in gene therapy has been the treatment of cancer. Approaches have includedtransferring cytokine genes to stimulate an antitumor immune response, delivering genes thatexpress prodrugs to tumors, and transferring genes to protect stem cells during high-dosechemotherapy. This review will focus on restoring the function of tumor suppressor genes. In thecontext of cancer, transient gene expression that triggers cancer cell death may be enough tomediate a therapeutic effect. Expression of the transgene in only a fraction of tumor cells also maynot be limiting because these cells may alter the growth of adjacent cells.[3]

Genetic Basis of Carcinogenesis

The gene families implicated in carcinogenesis include dominant oncogenes like ras and tumorsuppressor genes like p53.[4-6] Proto-oncogenes (normal counterparts of oncogenes) ordinarilyparticipate in such functions as signal transduction (relaying information from the outer cellmembrane to the nucleus) and gene transcription. An abnormality (point mutation, amplification,translocation, or rearrangement) in only one of the two alleles of a proto-oncogene is sufficient toconvert it into an active oncogene, that is, to dysregulate its function and lead to malignanttransformation.In contrast, a single normal tumor suppressor gene in a cell is typically sufficient to perform thenormal function of the gene, but loss of function of both alleles, by mutation or deletion or acombination thereof, leads to dysregulation of cellular growth. In addition, some tumor suppressorgene mutations�for example, some p53 mutations�act in a �dominant negative� manner, that is, amutation in one allele can lead to production of a mutant p53 protein that binds to, and therebyinactivates, the structurally normal protein encoded by the opposite allele. Viral proteins also maybind and functionally inactivate p53.[7]

Page 1 of 9

Page 2: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

Rationale for Restoring p53 Function in the Cancer Cell

All cancers are thought to contain multiple abnormalities in a variety of genes that control variousaspects of cell growth and development. Thus, correcting all the genetic abnormalities in the cancercell might seem necessary to reverse the malignant process. Correction of all genetic abnormalitieswould be an impossible task, however, particularly since some of these abnormalities have not yetbeen identified. Moreover, individual patterns of expression would need to be assessed�for eachgene in every patient. Fortunately, correcting a single genetic abnormality is enough in some casesto induce tumor cell death by apoptosis.[8-10]A number of in vitro studies that used cultured cancer cell lines demonstrated that eliminating theexpression of a single dominant oncogene (ras) or adding a normal copy of a tumor suppressor gene(p53 or the retinoblastoma [Rb] gene) to cells that had deleted or mutated copies of these genesreduced or even abolished critical aspects of the malignant phenotype, such as tumorigenicity inanimals or anchorage-independent growth.[10-12] Because of this, the problem of simultaneouslycorrecting multiple functional genetic defects in the cancer cell did not appear as daunting.Restoring normal gene function to every cancer cell, which is beyond the capabilities of the vectorscurrently available for use in gene therapy, was also thought to be necessary at one time. However,transduced cells expressing a toxic transgene are now recognized to alter the growth of adjacentnontransduced cells. This has been termed the bystander effect.[3]While multiple genes offer potential targets for gene therapy in several common malignancies, ourgroup has focused on the goal of replacing normal p53 function. Many of the identified tumorsuppressor genes and proto-oncogenes encode proteins that are components of a network thatconverges on the protein encoded by the Rb gene.[13,14] Phosphorylation of the Rb proteinincreases transcription of other genes and protein synthesis, leading to cell growth. Phosphorylationof Rb is controlled by a multimolecular complex of proteins, containing cyclins and cyclin-dependentkinases as well as a potent inhibitor of most cyclin-dependent kinases, termed p21.[15,16] In turn,one of the many functions of the p53 protein is regulating p21 function.[17,18] Thus, p53 plays acentral role in regulating the cell cycle because it indirectly regulates the function of Rb. When p53function is normal, this pathway is tightly regulated; however, when p53 mutates or is absent,uncontrolled cell growth reflects lost control of the pathway.Moreover, p53 plays a central role in other metabolic pathways, including, importantly, control ofapoptosis.[19] In response to various toxic insults to cells, such as exposure to ionizing radiation orchemotherapy, normal cells either pause in their cell cycle long enough to repair DNA damage or, inother cases, undergo apoptosis; repair and programmed cell death both prevent the damaged DNAfrom being passed along to the cell�s progeny. When normal p53 function is absent, however,damaged DNA is much more likely to be passed along.[20]

Selecting the Tumor Model

Non�small-cell lung cancer is a logical target for novel therapeutic strategies for several reasons. It isthe leading cause of cancer death in the United States, and the median survival of patients whopresent with stage III or IV disease, as the majority of patients do, is measured in months. For mostpatients who are not potentially curable by surgery, other standard therapies are relativelyineffective. Radiotherapy, which offers the best chance for locoregional control of disease in patientswith surgically unresectable disease, is successful in only 20% of cases, and local primary failure orrecurrence may be the only site of failure in up to one third of patients.[21]Mutation or inactivation of p53 occurs in a high proportion of nearly all common human cancers,including non�small-cell lung cancer.[21-23] In view of the key role of p53 in cell-cycle regulationand apoptosis and the role of defective p53 function in carcinogenesis, attempting to replace p53represents a logical gene replacement strategy. My colleagues and I have therefore studied thisapproach in patients with non�small-cell lung cancer. Our initial approach has been based oninjecting the primary tumor with a vector expressing wild-type p53, with the aim of improving thelocoregional control of non�small-cell lung cancer.

Selecting an Efficient Vector

Most available vectors were inefficient in transducing genes into cancer cells. Thus, the nextexperimental goal was to develop an efficient vector to deliver a therapeutic gene into cancer cells.Studies of models of human tumors in vitro and in nude (immunoincompetent) mice showed thatuptake of retroviral vectors that contained wild-type p53 or antisense ras were efficient enough to

Page 2 of 9

Page 3: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

mediate a therapeutic effect. In contrast, control vectors carrying cancer-associated p53 mutationsdid not suppress cell growth, indicating that the therapeutic effect was a direct result of normal p53expression and not a nonspecific effect caused by transduction of the vector or the p53sequences.[24]These experiments and many other early studies, including our first clinical trial of p53 genereplacement therapy, used a retroviral vector that contained wild-type p53 cDNA linked to a b-actinpromoter. While retroviral vectors yield levels of gene expression sufficient to demonstrate a biologiceffect, vectors derived from adenoviruses can achieve much higher levels of gene expression, whichshould offer greater therapeutic potential. Further, adenovirus vectors have the advantage ofinfecting both dividing and nondividing cells, in contrast to retroviral vectors, which infect onlyactively dividing cells. Adenovirus vectors do not integrate into the genome, so expression istransient. This is not necessarily a disadvantage in cancer patients, however, since prolongedexpression is not required or even necessarily desirable after the tumor cell kill.Given these theoretic advantages of adenoviral vectors, we selected first-generation adenoviruses inwhich nonessential genes were deleted and replaced with wild-type p53 driven by a cytomegaloviruspromoter (ie, adenovirus p53 [Ad-p53]). In animal studies, subsequent treatment with Ad-p53 greatlyinhibited the tumorigenicity of intratracheally inoculated lung cancer cells.[25]

Preclinical Studies of p53 Gene Therapy

The demonstration of an antitumor effect for normal p53 transduced into cancer cells growing inculture notwithstanding, we were concerned that only a fraction of tumor cells would be transducedin vivo, limiting therapeutic efficacy. Thus, the discovery that viral vectors can readily penetrate3-dimensional cancer cell matrices, indicating that they would spread beyond the site of intratumoralinjection, was encouraging.[26] Furthermore, adenovirus that contains wild-type p53 is not toxic tonormal bronchial epithelial cells in culture, and it is minimally toxic in mice, even at very high levels,suggesting a very favorable safety profile for clinical trials.[27] In addition to the work reported withnon�small-cell lung cancer, studies in nude mouse models of squamous cell carcinoma of the headand neck have demonstrated a significant therapeutic effect for adenovirus p53, confirminginduction of significant apoptosis.[28]Despite the high efficiency of the cancer cell transduction achieved by viral vectors and the spreadof injected vectors beyond the site of intratumoral injection, it is unlikely that every tumor cell will betransduced. In vivo studies of intratumoral injections of Ad-p53 into subcutaneous tumors in miceshowed a degree of regression exceeding that predicted by the transduction efficiency of the vector,suggesting bystander effects. This phenomenon, which is critical to achieving a widespreadantitumor effect, was first noted in vivo in brain tumor cells transduced with the herpes simplexthymidine kinase gene and then exposed to ganciclovir (Cytovene). The ganciclovir is nontoxic tonormal cells, but the transduced viral thymidine kinase converts the drug into cytotoxic ganciclovirtriphosphates, leading to the death of the transduced tumor cells.[3]In contemplating gene replacement therapy with p53 or other tumor suppressor genes,demonstrating that a bystander effect similar to that seen with the thymidine kinase gene alsooccurred in the case of p53, was essential. Using in vivo mixing experiments, investigatorsdemonstrated that retroviral wild-type p53-transduced cells could reduce the growth rate ofnontransduced human lung cancer cells, indicating an operative bystander effect in this system.[29]Several mechanisms may mediate bystander effects, which may differ depending on the genetransduced and the specific type of tumor cell. These mechanisms include transfer of toxic metabolicproducts through gap junctions, phagocytosis of apoptotic vesicles of dead tumor cells by live tumorcells that mediate apoptosis, induction of an immune response against the tumor, and inhibition ofangiogenesis.The so-called death receptor, Fas, also might mediate a bystander antitumor effect followingtransduction of a p53 expression vector. For example, H358 lung cancer cells express abundant Fasligand but not the Fas protein needed for apoptosis. Wild-type p53 upregulates expression of Fasprotein by these cells, and because the protein is released in soluble form, it may traffic to othercells, leading to a bystander effect.[30]Expression of the wild-type p53 gene also may downregulate the expression of vascular endothelialgrowth factor, which is one of the most common factors stimulating angiogenesis in human cancers.This downregulation reduces the formation of blood vessels by tumor cells, thereby possiblyinhibiting the continued growth of the cancer.In another study, the growth of lung cancer was studied using an orthotopic experimental model in

Page 3 of 9

Page 4: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

which human lung cancer cells grew as xenograft tumors in nu/nu mice. Directly administering aretroviral wild-type p53 expression vector into the tumor cells in vivo suppressed tumor growth.[31]Subsequently, we used an adenovirus expression vector to deliver wild-type human p53 cDNA totumor cells. This p53 expression vector induced apoptosis in cancer cells carrying a mutated ordeleted p53, but had little effect on the growth of cells containing wild-type p53. Similarp53-adenovirus vector constructs also inhibited growth of rat gliomas, human head and neckcancers, and human colon cancers in nude mice and can mediate p53 gene expression in bladderand liver cancers.[32] The products of other tumor suppressor genes, such as p16 and a truncatedRb gene also have been found to suppress tumor growth in animal models.[32]

Synergistic Effect of Transduced p53 With Other Therapies

In addition to the antitumor effect of transduced tumor suppressor genes used as single modalities invarious experimental systems, preclinical studies demonstrated synergy between p53 replacementtherapy and DNA-damaging chemotherapeutic agents that are useful against non�small-cell lungcancer, such as cisplatin and etoposide (VePesid).[33] Chemotherapy enhances expression oftransduced genes, whether viral or nonviral vectors are used, with a range of promoters. In onemodel, adding cisplatin before p53 transfection increased apoptosis and suppression of tumorgrowth.[34] The mechanism underlying this effect is not yet known. Preclinical studies also haveindicated that gene therapy may increase sensitivity to radiation.We tested whether adenoviral-mediated wild-type p53 gene expression sensitizes colorectal cancercells to ionizing radiation.[35] Wild-type p53 gene transfer into the SW620 colorectal carcinoma cellline was performed using Ad-p53 to evaluate the effect of wild-type p53 expression on radiationsensitivity. Based on the fact that survival at 2 Gy was reduced from 55% to 23% (Figure 1), theresults indicated that this vector sensitized the cells. Flow cytometric analysis of terminaldeoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay-labeled cells andin situ TUNEL staining of xenograft tumors demonstrated an increase in labeled cells after thecombination treatment. Compared with p53 gene therapy alone, this combination strategysignificantly enhanced the suppression of tumor growth in an animal model of subcutaneous tumor.The delay in regrowth to control tumor size of 1,000 mm³ was 2 days for 5 Gy, 15 days for Ad-p53,and 37 days for Ad-p53 plus 5 Gy, indicating synergistic interactions. These data indicate that thedelivery of wild-type p53 to cells with p53 mutations increases their radiation-sensitivity and that thisdelivery may be accomplished by adenoviral-mediated gene therapy.Additional experiments were conducted to ascertain whether the Ad-p53 vector also would sensitizenon�small-cell lung cancer cells to radiation. Two different non�small-cell lung cancer lines wereexamined, H358 and H1299. Both of these cell lines have homozygous deletions of the p53 gene.The H358 line was examined in vitro. In this case, the cells growing in culture were infected withAd-p53 at a multiplicity of infection (number of infectious viral particles per tumor cell) of 70 and thecells were irradiated 48 hours following infection. Sensitization was assessed using an in vitroclonogenic assay. Survival at 2 Gy was reduced from 70% for the cells receiving 2 Gy alone to 50%for the cells receiving both treatments. The H1299 cells were injected into the hind legs of nudemice. When the tumors reached 6 to 8 mm in diameter, they were injected with 1.5 × 1010 viralparticles. Two days later, the xenograft tumors were given a single radiation dose of 5 Gy. Similar tothe results with the colorectal xenografts, the non�small-cell lung cancer xenograft tumors had adramatic response to the combined treatment (Figure 2). The delay in regrowth to a control tumorsize of 1,000 mm³ was 1 day for the 5-Gy dose alone, 14 days for the Ad-p53 alone, and 36 days forthe combination of Ad-p53 plus 5-Gy treatments. Thus, the results for the combination of virus andradiation appeared to indicate a synergistic interaction and validate the use of this gene therapystrategy to treat non�small-cell lung cancer.Based on the results of recently completed preclinical studies, Ad-p53 in combination with radiationalso may be a useful strategy for treating brain tumors. To determine the effects ofadenovirus-mediated delivery of wild-type p53 on the radioresponse of glioma tumor cells, in vitroexperiments were performed.[36] The responses of two human glioma cell lines were compared:U87MG, which has wild-type p53, and U251MG, which has a mutant p53 allele. Monolayer cultures ofthese cell lines were infected with Ad-p53, control vector dl312 or culture medium. The cultures wereirradiated 2 days later and colony formation efficiency determined.Transfection with p53 had only a minor effect on the plating efficiency of unirradiated U87MG cellsbut significantly enhanced the radiosensitivity of these cells. The surviving fraction at 2 Gy wasreduced from 0.61 in controls to 0.38 in p53-transfected U87MG cells (Figure 3). The control vector,

Page 4 of 9

Page 5: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

dl312, did not influence radiosensitivity. The Ad-p53 vector was toxic to the U251MG cell line and,therefore, its influence on radiosensitivity could not be determined. A flow cytometric analysis ofTUNEL-stained cells demonstrated that the Ad-p53 vector sensitized the U87MG cells toradiation-induced apoptosis; the percentage of TUNEL-positive cells was increased following 9 Gyfrom about 3% in the controls to 19% in the Ad-p53 plus 9-Gy�treated cells. These data suggest thatadenovirus-mediated p53 may enhance the radioresponse of brain tumor cells with wild-type p53and that this radiosensitization may at least partially involve a restoration of the propensity forapoptosis. Additional support for this concept is derived from a study showing that radiationimproves immediate transduction efficiency and duration of transgene expression from anadenoviral vector.[37]

Clinical Studies

In our initial human study, nine patients with non-small-cell lung cancer received a retroviral vectorcontaining the wild-type p53 gene under the control of a b-actin promoter.[38] Other treatments hadfailed in these patients, all of whose tumors had been documented as having a p53 mutation. Sincetransduction via retrovirus was known to yield low titers of the transduced gene, the vector wasinjected into the tumor on 5 consecutive days, using either a bronchoscope or, in the case of chestwall lesions, a percutaneous needle.Regression of the injected lesion was observed in three patients, while disease stabilized in threeothers. One patient who died of a progressive kidney metastasis showed no evidence of viable tumorat the treated site at autopsy 4 months following the injection. One patient�s primary diseaseprogressed, and two patients were inevaluable (one could not tolerate the general anesthesia and sodid not complete treatment, and the other died within 3 weeks of treatment). Polymerase chainreaction (PCR) and/or in situ hybridization studies of posttreatment biopsies from eight patientsbefore treatment effects were evaluated showed that tumor cells had indeed integrated vector DNAsequences in up to 20% of cells in certain areas of the tumors. An important finding was that TUNELstaining (which detects DNA nicking) of the posttreatment biopsy samples showed that apoptosishad increased following treatment compared with the pretreatment baseline.No side effects attributable to the p53-vector sequences occurred in any patient, althoughbronchoscopy-related complications occurred in three patients. We found no evidence of retroviralsequences in DNA extracted from lymphocytes, sputum samples, or various nontumor tissuesobtained at autopsy on three patients. These results confirmed a high safety profile, successfuldelivery of normal p53 sequences to tumor cells, and a biologic effect of the transduced gene.These promising results have been supported by a more recent two-arm study in which a wild-typep53 gene was given to 52 patients with non�small-cell lung cancer using an adenovirus vector.Patients received Ad-p53 either alone or preceded by cisplatin, 80 mg/m² over 2 hours, 3 days beforep53 injection (cisplatin was chosen because of preclinical evidence of a synergistic effect with p53gene replacement). A single intratumoral injection of p53 was given, either bronchoscopically orguided by computed tomography, once per month for up to 6 months. Most patients had receivedprevious chemotherapy, in some cases with cisplatin, and all had tumor progression duringconventional therapy before entry into the study.As with the study using the retroviral vector, both clinical and laboratory evidence of p53 expressionwere seen. Ad-p53 alone (26 evaluable patients) mediated two partial responses and stabilizeddisease in 16 patients. A higher dose of Ad-p53 increased progression-free survival. Ad-p53 pluscisplatin (23 evaluable patients) mediated a partial response in two patients who had been treatedwith cisplatin previously. One additional patient achieved a partial response but did not have therequired follow-up documentation to confirm the response. Ad-p53 plus cisplatin prolongedprogression-free survival compared with Ad-p53 alone. A majority of patients also had evidence ofvector DNA by PCR of posttreatment biopsies. Although antiadenovirus antibodies were detected inall patients after one treatment, no anaphylaxis or other toxicities except transient feveraccompanied subsequent treatments. Perhaps surprisingly, despite high levels of serumantiadenovirus antibody, p53 transgene expression occurred in the tumor cells, and clinicalresponses were maintained.Although these studies corrected only one of the many genetic abnormalities present innon�small-cell lung cancer, evidence suggests an antitumor effect based directly on p53-mediatedapoptosis, as shown in the retroviral p53 trial. The two trials provided no evidence for the contraryview that tumor stabilization and regression resulted from a nonspecific immune reaction. In theAd-p53 study, PCR detected vector DNA in the majority of patients, but fewer patients showed

Page 5 of 9

Page 6: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

evidence of gene expression. Detection of gene expression following transfer of wild-type p53 in vivois difficult because successful transfer and expression of wild-type p53 in a tumor may destroyevidence of gene expression if apoptosis is induced and the cells die. However, in those patientswhose serial gene expression could be quantitated by immunohistochemistry, it was clear thatexpression of the transgene occurred despite the presence of high titers of antiadenovirus antibody.It is possible that serum antibodies have little effect due to poor penetration of solid tumors as aresult of high interstitial fluid pressure. Whether suppressing the antiadenovirus immune responsewould further enhance levels of transgene expression in the tumor is not known. In any case,repeated intratumoral injections of adenoviral p53 appear safe despite increases in antiadenovirusantibodies.A similar strategy has been reported for treating recurrent head and neck squamous cellcarcinomas.[39] Head and neck cancer patients may benefit from improved local control, as theprincipal cause of death for these patients is locoregional recurrence. In a phase I dose-escalationstudy, 33 patients were injected with doses of up to 1011 plaque-forming units. Dose-limiting toxicityor serious adverse events were not observed. Gene expression in tumor tissue was documented.Two of the 17 patients who had unresectable disease had a partial response and six had stabledisease for up to 3.5 months. One of the patients with resectable disease was noted to have apathologic complete response at the time of resection. Infectious Ad-p53 was detected in sputumand urine at the highest doses of 1011 infectious particles. Interestingly, the excretion time coursedid not change in duration or amount with subsequent injections despite the presence of high titersof antiadenovirus antibody. This raises the intriguing possibility that the antibody may not limit thebioavailability of systemically administered Ad-p53.

Future Research and Conclusions

The first patient treated with retroviral p53 received general anesthesia, but outpatient treatment isnow given using only topical anesthesia. Injection takes about 20 minutes. The simplicity of thetreatment procedure suggests that gene transfer using Ad-p53 may provide highly cost-effectivetherapy. Given the promising results of these phase I studies, phase II trials in patients withnon�small-cell lung cancer or head and neck cancer are now in progress, and plans also are underway to expand these studies to include patients with locally recurrent breast, bladder, and ovariantumors who may benefit from more aggressive treatment of locoregional disease. Based on thepromising preclinical data indicating that Ad-p53 and ionizing radiation are synergistic in causingcancer cell apoptosis, a phase II trial has been initiated in patients with unresectable, untreatednon�small-cell lung cancer combining Ad-p53 given by intratumoral injection with external-beamionizing radiation therapy.The low viral titer achieved with the retroviral p53 vector and its inhibition by circulatingcomplement make it unsuitable for systemic (rather than intratumoral) use. These limitations do notapply to the adenovirus vector, but its use systemically would require enhanced transgeneexpression, low vector toxicity, and, ideally, elimination of the immune response to the vector.Modification of the vectors used may reduce expression of endogenous adenoviral proteins, leadingto a diminished anti-adenoviral immune response.[40] The addition of agents that enhancetransgene expression may increase the effectiveness of low levels of transgene expressionsufficiently to render them therapeutic.Also useful would be a method of targeting delivery to the tumor. Early clinical work has suggestedthat some systemic effect occurs from adenovirus shed from the intratumoral injection site, asautopsy evidence has indicated that vector DNA was detected in distant tumor deposits. Other areasof current research include methods of increasing levels of p53 expression and suppression of theimmune response via concomitant administration of low-dose etoposide.[32]In conclusion, clinical trials have shown that repeated intratumoral administration of Ad-p53 is welltolerated and not associated with major toxicity. Evidence supports both gene transfer and geneexpression in the majority of the tumors. Progressively growing tumors refractory to conventionaltreatments have shown objective clinical responses to Ad-p53.In the near future, potentially promising areas for gene replacement therapy include the treatmentof locally advanced disease, adjuvant and neoadjuvant treatment of patients with resectable tumors,the use of gene therapy to sensitize tumors to radiotherapy and chemotherapy, and, potentially, thetreatment of localized premalignancy. Gene replacement therapy also may come full circle sincebasic and clinical research in cancer also may help solve the obstacles that have inhibited theapplication of gene therapy to monogenic inherited diseases. Given the rapid pace of recent

Page 6 of 9

Page 7: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

progress and the explosion of interest in gene therapy, we envision that in the near future, genetherapy will take its place alongside chemotherapy, radiotherapy, and surgery as one of the toolsroutinely available to help treat patients with cancer. References: 1. Anderson WF: Human gene therapy. Science 256:808-813, 1992.

2. Blaese RM, Culver KW, Miller AD, et al: T lymphocyte-directed gene therapy for ADA-SCID: Initialtrial results after 4 years. Science 270:475-480, 1995.

3. Culver KW, Ram Z, Wallbridge S, et al: In vivo gene transfer with retroviral vector-producer cellsfor treatment of experimental brain tumors. Science 256:1550-1552, 1992.

4. Bishop JM: Molecular themes in oncogenesis. Cell 64:235-248, 1991.

5. Weinberg RA: Tumor suppressor genes. Science 254:1138-1146, 1991.

6. Weinberg RA: Oncogenes and tumor suppressor genes. CA Cancer J Clin 44:160-170, 1994.

7. Werness BA, Levine AJ, Howley PM: Association of human papillomavirus types 16 and 18 E6proteins with p53. Science 248:76-79, 1990.

8. Baker SJ, Markowitz S, Fearon ER, et al: Suppression of human colorectal carcinoma cell growth bywild-type p53. Science 249:912-915, 1990.

9. Mukhopadhyay T, Cavender A, Tainsky M, et al: Expression of antisense k-ras message in a humanlung cancer cell line with a spontaneous activated k-ras oncogene alters the transformed phenotype(abstract 1799). Proc Am Assoc Cancer Res 31:304-304, 1990.

10. Takahashi T, Carbone D, Takahashi T, et al: Wild-type but not mutant p53 suppresses the growthof human lung cancer cells bearing multiple genetic lesions. Cancer Res 52:2340-2343, 1992.

11. Bookstein R, Shew JY, Chen PL, et al: Suppression of tumorigenicity of human prostate carcinomacells by replacing a mutated RB gene. Science 247:712-715, 1990.

12. Alemany R, Ruan S, Kataoka M, et al: Growth inhibitory effect of anti-K-ras adenovirus on lungcancer cells. Cancer Gene Ther 3:296-301, 1996.

13. Chen PL, Scully P, Shew JY, et al: Phosphorylation of the retinoblastoma gene product ismodulated during the cell cycle and cellular differentiation. Cell 58:1193-1198, 1989.

14. Kaelin WG Jr, Pallas DC, DeCaprio JA, et al: Identification of cellular proteins that can interactspecifically with the T/E1A-binding region of the retinoblastoma gene product. Cell 64:521-532, 1991.

15. Eastham JA, Hall SJ, Sehgal I, et al: In vivo gene therapy with p53 or p21 adenovirus for prostatecancer. Cancer Res 55:5151-5155, 1995.

16. Harper JW, Adami GR, Wei N, et al: The p21 Cdk-interacting protein Cip1 is a potent inhibitor ofG1 cyclin-dependent kinases. Cell 75:805-816, 1993.

17. el-Deiry WS, Tokino T, Velculescu VE, et al: WAF1, a potential mediator of p53 tumorsuppression. Cell 75:817-825, 1993.

18. Polyak K, Waldman T, He TC, et al: Genetic determinants of p53-induced apoptosis and growtharrest. Genes Dev 10:1945-1952, 1996.

19. Yonish-Rouach E, Resnitzky D, Lotem J, et al: Wild-type p53 induces apoptosis of myeloidleukaemic cells that is inhibited by interleukin-6. Nature 352:345-347, 1991.

Page 7 of 9

Page 8: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

20. Harris CC: The p53 tumor suppressor gene as a target for new anticancer therapies. Adv Oncol14:3-10, 1998.

21. Schaake-Koning C, van den Bogaert W, Dalesio O, et al: Effects of concomitant cisplatin andradiotherapy on inoperable non�small-cell lung cancer. N Engl J Med 326:524-530, 1992.

22. Chiba I, Takahashi T, Nau MM, et al: Mutations in the p53 gene are frequent in primary, resectednon�small-cell lung cancer: Lung Cancer Study Group. Oncogene 5:1603-1610, 1990.

23. Harris CC: The 1995 Walter Hubert lecture�Molecular epidemiology of human cancer: Insightsfrom the mutational analysis of the p53 tumour-suppressor gene. Br J Cancer 73:261-269, 1996.

24. Cai DW, Mukhopadhyay T, Liu Y, et al: Stable expression of the wild-type p53 gene in human lungcancer cells after retrovirus-mediated gene transfer. Hum Gene Ther 4:617-624, 1993.

25. Zhang WW, Fang X, Mazur W, et al: High-efficiency gene transfer and high-level expression ofwild-type p53 in human lung cancer cells mediated by recombinant adenovirus. Cancer Gene Ther1:5-13, 1994.

26. Fujiwara T, Grimm EA, Mukhopadhyay T, et al: A retroviral wild-type p53 expression vectorpenetrates human lung cancer spheroids and inhibits growth by inducing apoptosis. Cancer Res53:4129-4133, 1993.

27. Zhang WW, Alemany R, Wang J, et al: Safety evaluation of Ad5CMV-p53 in vitro and in vivo. HumGene Ther 6:155-164, 1995.

28. Clayman GL, Liu TJ, Overholt SM, et al: Gene therapy for head and neck cancer: Comparing thetumor suppressor gene p53 and a cell cycle regulator WAF1/CIP1 (p21). Arch Otolaryngol Head NeckSurg 122:489-493, 1996.

29. Nishizaki M, Fujiwara T, Tanida T, et al: Recombinant adenovirus expressing wild-type p53 isantiangiogenic: A proposed mechanism for bystander effects. Clin Cancer Res 5:1015-1023, 1999.

30. Owen-Schaub LB, Zhang W, Cusack JC, et al: Wild-type human p53 and a temperature-sensitivemutant induce Fas/APO-1 expression. Mol Cell Biol 15:3032-3040, 1995.

31. Fujiwara T, Cai DW, Georges RN, et al: Therapeutic effect of a retroviral wild-type p53 expressionvector in an orthotopic lung cancer model. J Natl Cancer Inst 86:1458-1462, 1994.

32. Roth JA, Cristiano RJ: Gene therapy for cancer: What have we done and where are we going? JNatl Cancer Inst 89:21-39, 1997.

33. Fujiwara T, Grimm EA, Mukhopadhyay T, et al: Induction of chemosensitivity in human lungcancer cells in vivo by adenovirus-mediated transfer of the wild-type p53 gene. Cancer Res54:2287-2291, 1994.

34. Nguyen DM, Spitz FR, Yen N, et al: Gene therapy for lung cancer: Enhancement of tumorsuppression by a combination of sequential systemic cisplatin and adenovirus-mediated p53 genetransfer. J Thorac Cardiovasc Surg 112:1372-1377, 1996.

35. Spitz FR, Nguyen D, Skibber JM, et al: Adenoviral-mediated wild-type p53 gene expressionsensitizes colorectal cancer cells to ionizing radiation. Clin Cancer Res 2:1665-1671, 1996.

36. Lang FF, Yung WK, Raju U, et al: Enhancement of radiosensitivity of wild-type p53 human gliomacells by adenovirus-mediated delivery of the p53 gene. J Neurosurg 89:125-132, 1998.

37. Stevens CW, Zeng M, Cerniglia GJ: Ionizing radiation greatly improves gene transfer efficiency inmammalian cells. Hum Gene Ther 7:1727-1734, 1996.

Page 8 of 9

Page 9: p53 Tumor Suppressor Gene Therapy for Cancer · 2017-10-19 · restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like

p53 Tumor Suppressor Gene Therapy for CancerPublished on Patient Care Online (http://www.patientcareonline.com)

38. Roth JA, Nguyen D, Lawrence DD, et al: Retrovirus-mediated wild-type p53 gene transfer totumors of patients with lung cancer. Nat Med 2:985-991, 1996.

39. Clayman GL, el-Naggar AK, Lippman SM, et al: Adenovirus-mediated p53 gene transfer inpatients with advanced recurrent head and neck squamous cell carcinoma. J Clin Oncol16:2221-2232, 1998.

40. Kovesdi I, Brough DE, Bruder JT, et al: Adenoviral vectors for gene transfer. Curr Opin Biotechnol8:583-589, 1997.

Source URL: http://www.patientcareonline.com/p53-tumor-suppressor-gene-therapy-cancer

Links:[1] http://www.patientcareonline.com/review-article[2] http://www.patientcareonline.com/authors/jack-roth-md-facs[3] http://www.patientcareonline.com/authors/stephen-g-swisher-md[4] http://www.patientcareonline.com/authors/raymond-e-meyn-phd

Page 9 of 9