Cell death and cancer: replacement of apoptotic genes and ... - Nature

13 downloads 136 Views 115KB Size Report
Cell death accounts for the majority of cancer cell elim- ination.12 ... act primarily via the induction of apoptosis, as do chemo- ...... 1939–1946. 144 van Meir EG ...
Gene Therapy (1998) 5, 728–739  1998 Stockton Press All rights reserved 0969-7128/98 $12.00 http://www.stockton-press.co.uk/gt

REVIEW

Cell death and cancer: replacement of apoptotic genes and inactivation of death suppressor genes in therapy M Favrot1,2, J-L Coll1,2, N Louis1 and A Negoescu1

´ ´ ´ Lung Cancer Research Group, Institut Albert Bonniot, Faculte de Medecine, Universite Joseph Fourier, Grenoble; and 2Department ´ ´ of Tumor Biology, Centre Leon Berard, Lyon, France

1

This review provides a critical evaluation of the increasing use of gene therapy in the treatment of malignancies to induce active cell death (ACD, apoptosis). This approach is consistent with the notion that cancer is an anomalous accumulation of cells largely resulting from diminished cell death. The review details the main genes potentially useful for therapy. Among these, p53 has received the majority of the investigators’ attention and provided encouraging results. Even greater hope is offered by newly tried direct inducers of apoptosis, such as bax, bclXS and caspases. Another fruitful direction is the association of apoptosisinducing gene transfer with radio- and chemotherapy,

which are also inducers of ACD. There is a delicate balance between cell gain through mitosis and cell loss in neoplasia because spontaneous apoptosis is widely present in tumors. In fact, the tumor environment favors bystander cell killing which appears to be a fundamental mechanism insofar as the rate of observed cell mortality cannot be accounted for by the known methods of gene transduction with efficiencies far below 100%. We conclude that apoptosis offers a mainstream approach for cancer gene therapy since ACD is highly inducible and only limited gains in malignant cell apoptosis may displace tumors from growth to regression.

Keywords: apoptosis; cancer; gene therapy

Introduction Neoplasia has long been envisioned as a disease of cell proliferation, occurring through enhanced rates of tumor cell division. In fact, there are neoplastic cells which divide more slowly than their benign counterparts but do succeed in producing tumors because of prolonged lifespan. Therefore, a broader image has emerged, perceiving cancer as an anomalous accumulation of cells engendered both by increased proliferation and diminished cell death.1,2 Indeed, among the multiple genetic events associated with tumor development are activation of antiapoptotic oncogenes such as bcl2,3 and inactivation of apoptosis-inducing tumor suppressor genes including p53 (abnormal in more than half of all malignant cell types)4 and bax.5–7 The term apoptosis describes genetically driven, active cell death (ACD). Apoptotic cells undergo separation from their neighbors, membrane blebbing, chromatin condensation, nuclear breakdown into micronuclei and cytolysis into condensed apoptotic bodies.8,9 Yet, viewing cancer from this standpoint may engender an underestimation of tumor cell loss, as shown by cell loss factor determination. Cell production, determined by mitosis quantification, allows the calculation of the potential doubling time (Tp, ie the time in which the cell number would have doubled had there been no cell

´ Correspondence: M Favrot, GRCPVA, Institut Albert Bonniot, Faculte ´ ´ de Medecine, Universite Joseph Fourier, Domaine de la Merci, 38706 Grenoble, France

loss), whereas the actual doubling time (Ta) is given by direct measurements of tumors. These two parameters permit the calculation of the cell loss factor (CLF = (1 − Tp/Ta) × 100).10 In transplantable animal tumors selected for rapid growth CLF exceeds 60%; it is still higher in human cancers, such as melanoma or colorectal carcinoma, and can reach 99% in some pulmonary tumors. Moreover, CLF augments during tumor enlargement.11,12 Cell death accounts for the majority of cancer cell elimination.12 Malignant tumors frequently display necrosis, which is mostly the result of hypoxia. Though conspicuous histologically, necrosis does not satisfactorily account for most of the losses predicted on the basis of CLF.13,14 It is therefore likely that apoptosis, which exists in all tumors,15,16 is responsible for an important part of cell death in neoplasia. Due to the rapidity of the phenomenon (2–5 min for cell condensation and fragmentation, 3 h for apoptotic-body phagocytosis and digestion), strong ACD-induced cell losses are often based on less than 5% of simultaneously visible apoptotic cells within a tissue.17 Moreover, tumor apoptosis occurs unevenly, in bursts. These features presumably precluded the precise quantification of ACD contribution to tumor cell deletion.12,18 The factors responsible for the spontaneous occurrence of ACD in malignant tumors are not unequivocally established.2 They presumably consist of two mechanisms: on the one hand, cell population regulatory mechanisms originating in the host organism, such as mild ischemia, suppression of survival signals originating in nontumor cells or death signals provided by the immune system (the so-called ‘social controls on cell death’19); on the other hand, apoptosis initiated by

Apoptosis–based gene therapy of cancer M Favrot et al

processes intrinsic to malignant cells, as indicated by different rates of ACD found in similar tumors expressing different oncogenes. For example, cancer cells frequently contain overexpressed myc gene family members20 and an inactivated retinoblastoma (rb) gene.21 The Myc protein signals cellular proliferation; yet, in cells lacking other specific mitogenic stimuli Myc induces ACD.22 Analogously, the Rb protein can be either an apoptosis inducer23 or suppressor.24 Taken together, the above data make plausible two assumptions. First, that the margins between tumor growth and regression could be crossed by a relatively minor augmentation in the rate of cell death. Second, that apoptosis, by its intrinsic property of being highly inducible, may provide a therapeutic way to increase cell losses. Indeed, over the last 15 years, a general concept has emerged that all nonsurgical modalities of cancer therapy act primarily via the induction of apoptosis, as do chemotherapy and radiotherapy, but also immunotherapy or hormone ablation.1,25–28 Mechanisms involved in therapyinduced ACD are only partially elucidated.2,27 Yet, a general property which provides the therapeutic window appears to be related to the sensitivity to apoptosis induction, which is generally lower in normal, as opposed to neoplastic cells. Thus, surrounding tissues may pause and repair the damage produced by therapy while tumor cells die through apoptosis. It is currently unclear exactly what determines this ‘threshold’ of apoptosis initiation, probably because multiple factors are likely to modulate the set point.29 Gene therapy, the newest approach for cancer therapeutics, has focused on two broad goals: DNA constructs are transduced to potentiate existing therapy, such as radiotherapy, or to deliver more specifically existing therapy, such as chemotherapy via drug sensitivity genes or immunotherapy; alternatively, transgenes are introduced to revert the genetic profile of malignancy within tumor cells.30,31 The broad spectrum of genetic abnormalities in cell transformation made unexpected the results given by the latter approach. Indeed, it might have been supposed that, to be effective, all the genetic lesions in every tumor cell would have had to be corrected, which is unlikely since vectors available have efficiencies far below 100%. Yet, it appears that correction of a single essential gene abnormality within only a fraction of tumor cells can be sufficient to induce tumor repression. Moreover, both copies of a tumor suppressor gene must be generally inactivated to eradicate its function.32 Therefore, replacement of only one normal copy of the gene in cells with homozygous loss of function can restore tumor suppression.30 Research in this area has recently concentrated on the direct induction of apoptosis through replacement of tumor suppressor genes involved in the cell death process or inactivation of ACD-inhibiting genes. Using others’ and our experience in this domain we will provide a current and critical global evaluation of this field of application of gene therapy.

The main mechanisms of apoptosis in tumor cells suggest candidate genes for cancer therapy A model progressively emerged for apoptosis induction giving the central position in ACD regulation to the

members of the bcl2 gene family.3,33 The bcl2 gene was discovered in a B cell lymphoma where a chromosomal translocation moves it into juxtaposition with trancriptional enhancer elements of the immunoglobulin locus.34 However, as described in various solid tumors, transregulatory mechanisms can also be responsible for the high levels of Bcl2 protein.3,35 Bcl2 heterodimerizes with Bax, a protein of the same family sharing 21% homology with Bcl2. Bax can be considered as the main effector of apoptosis. Its function in active cell death as a homodimer Bax-Bax is opposed by heterodimerization with Bcl2 (Bax–Bcl2). This model was substantiated by results showing that Bax also heterodimerizes with other ACDinhibiting members of the Bcl2 family such as BclXL, Mcl1, A-1;36 it is also corroborated by the correlation between Bcl2 expression and both low spontaneous ACD in tumors and general resistance to cancer therapy.3,5,35,37,38 Conversely, Bax expression is correlated with high incidence of apoptosis in lung and stomach cancers5,39 and low Bax levels are associated with poor response to chemotherapy in breast and ovary tumors.6,7 As recently demonstrated, lack of Bax protein in cancer cells can be the result of mutations in the bax gene.40 BclX is another member of the bcl2 gene family. Due to alternate mRNA splicing, bclX is transcribed into long (bclXL) and short (bclXS) forms. The BclXL protein, like Bcl2, functions as an inhibitor of apoptosis. In contrast, the BclXS protein is a dominant inducer of ACD.41 However, BclXS acts as a monomer and does not show the ability to form heterodimers with other Bcl2 family members.42 The final step of apoptosis induction, termed ‘executioner’ by Martin and Green,43 is represented by proteases of the interleukin-1␤ converting enzyme (ICE) family, which directly generate cell destruction; they have recently been grouped under the term ‘caspases’ for ‘cysteine-aspartic acid specific proteinase’.44 Caspases are most probably stimulated by the Bax-Bax homodimer and the process can be opposed by heterodimerization with Bcl2 (Bax-Bcl2) producing caspase blocking and cell survival.45,46 Recent results indicate that Bax can induce apoptosis independently of dimerization processes47,48 and, furthermore, without caspase activation;49 caspaseindependent execution was also described for another death-inducing homologue of Bax, Bak.50 This would imply that Bax and Bak are not merely inducers of the executioner but, together with caspases, components of it. This model proved useful to explain tentatively the function of most ACD regulators, including those not belonging to the bcl2 family. The first among these is the P53 protein. The mechanisms underlying apoptosis induction by P53 are as yet obscure.51 The wild-type (wt) p53 gene is induced in response to genotoxic stresses, including DNA damage, oxygen deficiency and altered ribonucleotide pools.52 According to the hypothesis put forward by Lane,53 P53 blocks the cell cycle to allow DNA repair. If repair fails, P53 triggers apoptosis. Thus, tumor cells lacking normal P53 will accumulate mutations and chromosome rearrangements leading to selection of highly malignant clones. This ‘guardian of the genome’ model for P53 function,53 while far from representing a complete explanation of P53-dependent ACD, is supported by the lack of normal p53 in more than half of all tumor cell types.4 P53 being the main known downregulator of bcl2, its mutations could result in elevated

729

Apoptosis–based gene therapy of cancer M Favrot et al

730

production of Bcl2. In addition, P53 strongly induces the expression of bax.54–56 Thus, p53 appears to be an important regulator of apoptosis through the Bax-Bcl2 balance, and any type of p53 inactivation could abrogate cell death through Bax/Bcl2 dysbalance, as recently confirmed in vivo.57 Accordingly, the survival advantage conferred by the absence of normal P53 in malignant cells may explain their increased drug resistance.4,58,59 Similarly, radiation-induced ACD is P53-dependent and is inhibited by Bcl2 and BclXL.58–63 Finally, overexpression of the murine double minute 2 gene (mdm2), which inhibits P53-directed transcription of target genes,64,65 may induce a multiple drug resistance phenotype in tumor cells expressing wt p53,66 presumably by blocking p53 apoptotic function.67 Caspase activation can also occur directly, without mediation by Bax homodimers. Such a mechanism is best illustrated by the tumor necrosis factor receptor (TNFR) family members, such as Fas (CD95, specific for the Fas ligand) and TNFR1 (which binds TNF␣).68 Upon its activation, Fas recruits FADD (Fas-associated protein with death domain). Similarly, TRADD (TNFR1-associated death domain protein) binds to TNFR1 and connects it to FADD via interaction between death domains. Subsequently, FADD activates FLICE (FADD-like ICE, caspase 8) inducing apoptotic execution.69 Thus, the TNFR family may be a promising therapeutic target since its direct caspase activation is both quick and irreversible.69 This brief presentation of the main pathways of apoptosis for which tumor cells may be deficient, readily suggests candidates for therapy: ACD-inducing genes and antisense constructs aimed to block apoptosis repressors. However, ACD augmentation would be effective against a tumor only if transduced DNA does not enhance the replication rate. This caveat does not apply to p53, which induces either G1 arrest or apoptosis.21 The difficulty may appear within the bcl2 gene family since Bcl2 reduces cell proliferation, and Bax, while antagonizing the cell survival function of Bcl2, also reverts its antiproliferative activity.70,71 Few results are available to resolve this matter. However, the obstacle seems limited when compared with the high apoptotic rates which gene transduction can provide, enhanced, as shown below, by various mechanisms of bystander cell killing.

Wild-type p53-mediated anticancer strategies Due to its widespread alterations in malignant cells and its central position in both proliferation arrest and apoptosis induction, p53 has been the most investigated candidate for apoptotic gene replacement therapy methods. Results provided by these studies are often contradictory. In vitro, with some exceptions, such as neuroepithelioma or pancreatic cell lines, transduced p53 suppressed growth of tumor cells, as described for leukemia and lymphoma, osteosarcoma, melanoma, stomach, colon, ovary, uterus, breast, prostate, bladder, head and neck, and lung cancers.72–96 However, while unequivocally demonstrated in some of these cell types (such as lymphoma, uterine, head and neck carcinoma and osteosarcoma cells) apoptosis was absent from cells originating in pancreatic cancers.79,82–84,97–100 Glioblastoma cells gave contrasted results: no apoptosis80,101 or, at the opposite

extreme, ACD initiation.102,103 The same conflicting results were noted between p53-transfected colon carcinoma cells92,104 and for hepatocarcinoma cells.80,105 Lung tumor cells are one of the most investigated targets for the effects of wt p53 transfection, which contributed to the application to lung cancer of the first clinical administration of p53. In vitro, ACD induction by transfection of wild-type p53 proved variable in these cells. Thus, p53 did not induce in vitro apoptosis of H358 cells, a p53-null human non-small cell lung cancer (NSCLC) line despite a virus-mediated transfection efficiency superior to 80%,106,107 a result contested by Katayose et al108 and Qazilbash et al.96 Alternatively, Fujiwara et al109 reported apoptosis induction in spheroids formed by H322 cells (a human NSCLC line bearing mutated p53) whose growth was inhibited by retroviral wt p53 transfection. In a small cell lung cancer (SCLC) line (N417, mutated p53) wt p53 transfection did induce apoptosis.110 In vivo, transduced p53 generally induced tumor growth retardation in nude mice bearing tumors produced by glioblastoma, osteosarcoma, melanoma, breast, colon, head and neck, and human lung malignant cells. Apoptosis was specifically detected in these tumors. However, in similar conditions, human pancreatic cancer cells produced tumors growing independently of p53 administration and ACD induction was not observed. Prostate cancer cells (148-1PA line, p53-null) implanted in syngeneic mice produced tumors unresponsive to p53 transduction; however, in nude mice, another prostatic line (Tsu-pr1, mutated p53) gave tumors whose growth was strongly retarded by wt p53. The antitumoral effect is largely related to the number of implanted tumor cells and the time allotted for tumor growth before treatment. Thus, head and neck squamous carcinoma cells when implanted in low amounts (2.6 × 106) in nude mice and treated before visible nodule formation produced no tumors, whereas starting with twice as many cells, allowed to produce 1 cm diameter nodules, gave only a 50% reduction in tumor size due to p53 transfection. Analogously, only 25% of the nude mice inoculated intratracheally with 2 × 106 squamous lung cancer cells (H226Br line, mutated p53) developed tumors when intratracheally instilled with wt p53 3 days after cell administration.76,77,80,82–84,93,95,96,103,104,111–115 The above contradictory experimental results suggest several explanations. The first is offered by p53 genotype of the treated cells. Indeed, in most of the studies cited above, growth inhibition and cell death were observed in cell lines defective for the P53 protein (mutated or absent). Yet, Liu et al84 reported cytocidal effect of p53 in head and neck cancer cells although wild-type P53 was expressed. Reciprocally, ACD did not occur after wt p53 transfection in neuroepithelioma, colon or pancreatic cancer cells despite the lack of endogenous wt P53.75,82,116 Surprisingly, in vitro P53-induced ACD seems to occur only above a certain percentage of transfected cells. Thus, p53 adenovirus-transduced glioma cells responded by apoptosis only at transfection efficiencies superior to 40%. Growth arrest and apoptosis were not observed below this proportion of infected cells, though the original status of their p53 gene was defective.117 Another explanation may be the cellular level of transduced wt P53 which dictates the response of the cell. Low levels of P53 result in cycle arrest whereas high levels result in apoptosis;118–120 at still lower levels, insufficient

Apoptosis–based gene therapy of cancer M Favrot et al

to cause cell cycle arrest, P53 offers protection against apoptosis.121 Moreover, recent results indicate that cotransfection of wt p53 together with a fusion peptide corresponding to the carboxy-terminal part of P53 activates DNA binding of both wt and mutant P53 resulting in strong induction of apoptosis.122 In any event, with most gene transfer efficiencies below 100%, the antitumoral effect of p53 relies strongly on the ability of transduced cells to induce apoptosis around them, in parental cells.123 This phenomenon was termed ‘by-stander killing’; it consists in the induction of apoptosis and was mainly investigated for suicide genes, in particular, herpes simplex virus thymidine kinase (HSVtk).124,125 In vitro, the phenomenon relies on toxicmetabolite circulation between transfected and nontransfected cells via apoptotic bodies,126,127 gap junctions128 or by diffusion in the culture medium.129 These mechanisms have been proposed for P53-induced bystander apoptosis of H322 and H358 lung cancer cells in vitro.96,106 In vivo, bystander apoptosis may be favored by increased cell– cell interaction130 and by apoptotic-body phagocytosis, stronger in tissues than in culture.18,96,131 Furthermore, bystander killing is boosted in vivo by an immune component which also allows the rejection of distant metastases and resistance to further rechallenges with nontransfected tumor cells.132,133 However, the immune basis of this distant bystander effect is still uncertain and could possibly be the result of either natural killer cells or of a still unknown soluble factor when observed in severe combined immunodeficient (SCID) and nude mice.134–136 These data are strengthened by very recent results showing that therapy-resistant (clonogenic) neoplastic cells fail to produce tumors if surrounded by apoptotic cells. When surrounding cells are not dead, but only cyclearrested, they presumably provide a ‘feeder’ support to the clonogenic cells allowing them to proliferate.137 P53 acts far upstream in the apoptotic pathway so that many other genes are epistatic to it.138 Therefore, apoptosis can be both dependent on, and independent of, P53 status and function.139 Moreover, P53-mediated apoptosis can occur, in certain cell types, in the absence of P53-dependent transcriptional activation of P53-target genes.67,140 P53 is also known to be capable of associating with a plethora of cellular proteins,141 and it is thus plausible that it may exert an ACD regulatory function through many of these factors. For example, Fas/APO1 is a direct transcriptional target of P53, as determined in vitro on colon and NSCLC cell lines. A certain threshold for the effects of P53 on Fas expression might exist in the presence of mutant P53.142,143 Wt p53 is also able to reduce tumor growth and metastases through inhibition of angiogenesis. Wild-type p53transduced glioma cells secrete an effector of yet uncertain structure, termed GD-AIF (glioma-derived angiogenesis inhibitory factor), which strongly neutralizes the angiogenic signals produced by the parental cells, as well as those of basic fibroblast growth factor.144 Moreover, P53 stimulates the in vitro production of thrombospondin I by fibroblasts, a potent inhibitor of vascular development.145 Finally, mutant P53 potentiates the induction of vascular endothelial growth factor (VEGF), an activator of tumor neoangiogenesis.146 Indeed, p53 gene transfer induced growth retardation in a model of human breast cancer cells implanted into nude mice. No evidence of cytotoxicity was detected in treated tumors, but they

exhibited a reduced number of blood vessels.123 Therefore, wt p53 restoration may have therapeutic implications beyond the growth and apoptosis regulation in individual cells and the complex mechanisms involved in the antitumoral effect of P53 in vivo cannot be entirely extrapolated from in vitro results. The abundant experimental data concerning p53 transfection provided a sound basis for clinical applications. To our knowledge, eight clinical protocols have been proposed, involving 72 patients. The first clinical results of wt p53 transfection have been recently reported by Roth and co-workers,147,148 who obtained tumor regression in three patients and tumor stabilization in three others out of nine non-small cell lung cancer (NSCLC)-afflicted humans. The lesions were treated either by bronchoscopic or by percutaneous injections with a retroviral vector containing the p53 gene. ACD induction was strong in seven patients (10.6 ± 2.9 apoptotic cells, versus 1 ± 0.5 for pretreatment biopsies) independently of their response to therapy. The same authors also proposed a clinical protocol involving combined wt p53-antisense ras therapy for NSCLC: out of six patients, one gave a complete response; two, a partial response; and three exhibited stable disease. Habib and coworkers proposed a protocol for p53 transfection in colorectal liver metastases and another one for hepatocellular carcinoma: the first elicited no response; the second induced one complete response, two partial responses, and one minor response in four patients.30 Venook and colleagues proposed a protocol for liver carcinoma and Clayman and co-workers proposed a wt p53 protocol for head and neck squamous cell carcinoma: no data are available about responses obtained in the latter two protocols.30,149

Current evolutions and promising directions to potentiate apoptosis induction through genetic engineering Gene therapy-based enforcement of chemotherapy and radiotherapy As ACD progressively gains a central place within the goals of cancer gene therapy, new mechanisms are devised to increase apoptosis-inducing strategies. One of these mechanisms is P53-mediated sensitization to ACD induced by nongenetic means of cancer therapy, ie drugs and radiation.2,25 Radiation-induced apoptosis is P53-dependent, as demonstrated both in vitro and in vivo.60,63,150 And indeed, wt p53 expression strongly potentiates the ACD-based cytotoxicity of ionizing radiation, as demonstrated in vitro using colon carcinoma cells. Interestingly, wt P53 did not have by itself a cytotoxic effect on these cells which expressed endogenous mutant P53 and responded to wt p53 transfection by mere growth arrest. Similar results have been reported for glioblastoma cells. In vivo, combined p53 and radiation therapy reduced colorectal carcinoma growth within nude mice more efficiently than each of the two methods performed alone.116,151,152 In vitro, expression of wt p53 into cancer cells powerfully enhanced their sensitivity to apoptosis induced by drugs, such as platinum salts and topoisomerase II inhibitors. These results were essentially obtained for glioblastoma, liver and colon cancer cells.116,151,153 Accordingly, mdm2 antisense oligonucleotides, by liberating P53

731

Apoptosis–based gene therapy of cancer M Favrot et al

732

activity, increased the in vitro susceptibility of a glioblastoma cell line to cisplatin-induced apoptosis.66 In vivo, experiments performed in nude mice for lung cancer cells indicated that cisplatin plus p53 gene transfer strategy yielded significantly greater apoptosis and tumor growth retardation than wt p53 gene transfer alone. The timing was shown as critical: cisplatin administration simultaneous with or subsequent to p53 gene was less effective than cisplatin-first sequential treatment.107,154 However, important exceptions exist here too. Wild-type p53 gene transduction in a p53-null pancreatic carcinoma cell line did not change its sensitivity to four different types of anticancer drugs, including cisplatin, etoposide, 5-fluorouracil and paclitaxel.82 Moreover, normal p53 function protected fibroblasts against the anticancer agent paclitaxel; in this case, p53-deficient cells stalled in G2/M upon drug action and triggered apoptosis, whereas cells harboring wt p53 traversed through mitosis and escaped ACD.155 Analogously paradoxical results were reported by Fan et al156 for cisplatin and pentoxifilline in breast cancer cells. Furthermore, wt P53 stimulated the promoter of the multidrug resistance gene when transfected into p53-null H358 NSCLC cells as in ovarian or colon carcinoma cell lines.157 Results obtained in vitro and in vivo with combined cisplatin and p53 transfection in lung cancer cells form the basis for the clinical protocol recently proposed by Roth and co-workers.158 This protocol employs two strategies, the first one exploring the adenovirus vector-connected toxicities, and the second consisting of effective wt p53 and cisplatin administration in patients. To emphasize even more the complexity of the mechanisms involved in P53-mediated potentiation of chemotherapy, in vitro incubation of acute myelogenous leukemia cells (wt p53) with p53 antisense oligonucleotides resulted in a 90% reduction of the cell population. When the surviving cells were replated in the absence of the oligonucleotides, a continuing decline in cell number was recorded. Since preclinical trials demonstrated a very low toxicity of these oligonucleotides, a phase I clinical trial was conducted in 16 patients. Though no clinical complete response was obtained, leukemic cell growth was reduced and their sensitivity to apoptosis-inducing agents augmented. Measurement of p53 expression demonstrated a transient decline during the initial treatment with the antisense construct, followed by a four-fold increase. This led the authors to hypothesize that the transient inability of p53 to respond to the stimulatory messages given by drug-induced DNA damage eventually results in p53 overexpression driving cells into ACD.159,160

Cancer gene therapy based on final inductors of apoptosis Conflicting results obtained with p53, a multifunctional gene acting far upstream in the ACD pathway, led to targeting cancer gene therapy toward the final steps of apoptosis induction. Indeed, ACD-induction efficiency might be inversely proportional to the length of the downstream pathway between the function of the transfected gene and cell execution. One of the factors thus considered is Waf1 (Cip1, P21), a known mediator of P53-induced growth arrest and apoptosis.161–163 Waf1 is a cyclin-dependent kinase (cdk) inhibitor which controls cdk-mediated inactivation of Rb

through phosphorylation. Rb inactivation permits DNA synthesis and mitosis which explains the large number of tumors containing a mutated, or deleted rb gene.164 Rb also inhibits apoptosis, presumably because its antiproliferative function facilitates the growth arrest necessary to DNA repair. In the absence of Rb, damaged cells would apoptose after receiving the signal to proceed inappropriately through the cycle.165 However, variable results were obtained by transfecting waf1, because it proved able to act through three mechanisms, ie cell cycle arrest, apoptosis initiation or cell differentiation. In vitro, it initiated ACD in cell lines of osteosarcoma cells, but not in lung H358 cells, melanoma or renal carcinoma cells. In the two latter cell types, it produced morphological changes, such as an increased nuclear:cytoplasmic ratio, and increases in adherence, and growth arrest, consistent with a differentiated phenotype. Results for breast cancer cells are contradictory: cell cycle arrest without ACD or, on the contrary, general apoptosis induction. Interestingly, before ACD induction, the majority of the waf1-transfected breast carcinoma cells displayed a 50 to 100-fold increase in size and persisted as single giant cells for several days without division, while the remainder underwent nuclear division (multiple nuclei) but were unable to complete cytokinesis. Some of these differences may be attributed to the status of p53 because P53 mutants can variably counter one or other of the functions of Waf1. In nude mice, waf1 retarded tumor progression in prostate cancer, melanoma and renal carcinoma cell-induced tumors, despite the lack of detectable apoptosis.77,108,112,166–169 In vitro transduction of another cdk inhibitor, p16INK4 (inhibitor of cdk4), inhibited proliferation of NSCLC cell lines. It also induced ACD if overexpressed together with p53 in liver and colon tumor cell lines, in which p53 alone did not induce apoptosis. Apoptosis dependent upon P16 appeared to be connected to its ability to repress Rb synthesis. In vitro, tumors produced in nude mice with NSCLC lines grew slower after p16 transfection; for liver and colon carcinoma cell-induced tumors, some growth retardation was observed with p16, little effect was seen with p53, but strong inhibition of tumor growth was obtained with the combination of the two genes.170,171 BclXS efficiency as an antitumoral agent has been recently tested. In vitro, its expression consistently induced growth arrest via apoptosis induction in cell lines originating in tumors of breast, stomach, colon and neuroblastoma. In accordance with its probable dimerization-independent apoptotic action, BclXS was strongly efficient even in the presence of high levels of Bcl2 or BclXL. Moreover, BclXS potentiated chemotherapyinduced apoptosis in cultured breast cancer cells. In vivo, BclXS suppressed growth of mammary tumor cells implanted in nude mice via apoptosis induction. Antitumoral activity was greater than predicted from the percentage of transduced cells, which raises the possibility of a BclXS-mediated bystander apoptosis.172–175 Bax in vitro transduction of breast and ovarian cancer cells did not affect viability but increased chemo-, and radio-induced apoptosis.176,177 During the last year, we concentrated upon comparing therapeutic effects of p53, bax and bclXS, in H322 (mutated p53) and H358 (p53null) bronchioloalveolar carcinoma cells.178 These indicated an important advantage for bax in terms of cytotoxicity and terminal deoxynucleotidyl transferase-mediated

Apoptosis–based gene therapy of cancer M Favrot et al

dUTP nick end-labeling (TUNEL)-quantified apoptosis over both p53 and bclXS, the latter two providing almost identical effects. Therefore, it is conceivable that transfection-induced Bax expression acts by enriching the cellular pool of Bax-Bax homodimers, which readily turns on caspases and initiates the execution of the cell. Such a mechanism could be more immediate and thus more efficient than ACD induction by P53. This interpretation is supported by the difference we observed in Bax cytotoxicity between H322 cells (about 70% of Bax-expressing cells died) and H358 cells (more than 90% cytotoxicity). Indeed, blot experiments revealed undetectable levels of Bcl2 in H-358 cells as opposed to a notable expression of this protein in H322 cells. Since P53 regulates the BaxBcl2 balance by repressing bcl2 and transactivating bax,54,55 bax-p53 cotransfections would be expected to enhance the apoptotic effect of bax by favoring the liberation of Bax from heterodimers with endogenous Bcl2 and the accumulation of ACD-inducing Bax-Bax homodimers. We were not able to identify such a cytotoxic advantage for bax-p53 cotransfections, possibly because there was little enhancement to be gained beyond the strong effect of bax acting alone. Our results showed poor, if any, bystander killing and growth inhibition by transfected p53 or bax in vitro. Finally, work in progress in our laboratory indicates that the in vivo antitumoral effect of bax is still stronger than that of p53 and that bax delays in vivo tumor growth via bystander mechanisms. The first attempt of tumoricidal therapy based on caspase gene transfection reported that ICE (caspase 1)induced apoptosis in the majority of transduced glioma cells, both in vitro and in vivo, despite their lack of functional P53; yet, caspase-tolerant tumor cells subpopulations could be detected.179 Finally, caspase activation can be obtained through Fas gene transfection, as demonstrated in vitro for glioma cell lines. The Fas/APO1 cDNA expression vector dramatically enhanced cell surface expression of Fas and induced susceptibility to Fas antibody-mediated apoptosis.180 Similarly, TNF␣-transduced lymphoma T cell lines underwent apoptosis. The ACD process was mediated by the released TNF␣ in autocrine and paracrine fashion and promoted a strong bystander killing of parental cells. Interestingly, while leaving unchanged P53 and Bax levels (as expected from its action downstream of these proteins) TNF␣ down-regulated Bcl2 expression, showing that its apoptotic function is complex. In vivo, TNF␣ secreted by in vitro engineered cells implanted in syngeneic mice together with parental tumor cells, induced tumor regression after a short initial growth. However, the effect is likely to depend on immunity since immunosuppression of animals eliminated the ability of TNF␣transduced tumors to regress.181–183

Oncogene inactivation therapy Oncogene inactivation concerns primarily antisense therapy directed against genes which encode signal transduction proteins. These approaches have succeeded in reversing the neoplastic phenotype and dramatically reducing cell proliferation and tumorigenicity. The most extensive application of the methodology is anti-ras therapy.184–187 Anti-raf,188 anti-fos189 and anti-myc190 have also been tried. These antisense procedures generally do not induce apoptosis, though raf is an inhibitor of ACD, pre-

sumably via raf,191 and protects cells against chemotherapy-induced ACD.192 Bcr-abl antisense nucleotides initiate in vitro apoptosis of chronic myelogenous leukemia cells and render these cells susceptible to ACD produced by chemotherapeutic agents. Most likely, the BcrAbl oncoprotein provides an internal stimulus which renders these cells independent of ACD triggers. By reducing this stimulus, when growth factors are not sufficiently available or a cytotoxic signal is present, cells can undergo apoptosis.193–195 In vivo, chronic myelogenous leukemia evolution was strongly retarded and cure was achieved in about 50% of severe combined immunodeficient mice treated with a combination of bcr-abl antisense oligonucleotides and either myc antisense DNA or cytotoxic drugs; apoptosis was demonstrated in these cells.196,197 The main target for survival-gene inactivation was bcl2 and antisense strategies have been developed to lower bcl2 expression. In vitro, they potentiated spontaneous apoptosis and chemotherapy-induced ACD of lymphoma cell lines or of acute myeloid leukemia-patient-derived cells.198–201 In vivo, SCID mice inoculated with lymphoma cells treated with antisense bcl2 before inoculation failed to develop malignancy. The likely explanation is that, by down-regulating bcl2 (about 30% reduction as compared with control) in a cell that is heavily dependent on Bcl2 for survival, the cell is committed to ACD. Even if Bcl2 expression increases after this critical point the cell cannot recover. Thus, antisense-treated cells, although apparently viable when they are injected into the mice, are doomed and tumors do not result.202 Bcl2 antisense strategy formed the basis of a first clinical protocol for non-Hodgkin lymphoma.203 An 18-base antisense oligonucleotide was subcutaneously administered for 2 weeks to nine patients. In two patients, tumors regressed and in two others, the number of circulating lymphoma cells decreased. The eight patients received further chemotherapy, six of them answering by favorable evolution. Encouragingly, the oligonucleotide maximum-tolerated dose was not reached, doses used being just above those effective in animals; no toxic effects were noted, which permits higher doses to be used in the future. This is particularly promising since there was concern that bcl2 antisense might affect cells that normally contain high levels of this protein in connection to their long life span, eg memory B cells or neurons. Yet, these cell types apparently resisted, possibly due to other ACD inhibitors or to limited access of the nucleotides to the brain. Alternatively, enforced expression of anti-apoptotic genes can be used to diminish ACD-based myelosuppression during cancer chemotherapy. Indeed, in vitro experiments demonstrated that bcl2-transduced murine bone marrow cells resist apoptosis induced by both topoisomerase I and II inhibitor cytotoxic drugs.204 Similarly, in vitro experiments demonstrated that bclXL is a strong inhibitor of chemotherapy-induced apoptosis. In addition, this property spans over a wide variety of cells and anticancer agents, its expression conferring a multidrug resistance phenotype.205,206

The incidence of gene transfer techniques on apoptosis induction Despite the central position of apoptosis in the action of almost all gene therapy protocols for cancer, little infor-

733

Apoptosis–based gene therapy of cancer M Favrot et al

734

mation is available about the involvement of DNA transfer techniques in ACD initiation. Adenoviruses produce several proteins which are implied in apoptosis regulation in the host cell. The main participants are the early (E) viral proteins E1A, which induce apoptosis by both p53-dependent and p53-independent mechanisms and E4, which initiates ACD independently of p53 expression.207–209 The viral e1B gene encodes two products that inhibit apoptosis, a 55 kDa protein which inactivates P53 and a 19 kDa protein which acts downstream, analogously to Bcl2, to block Bax.210–213 Adenoviruses engineered for gene therapy have generally e1A and e1B deletions to preclude uncontrolled virus replication, but e4 has been mostly left functional and thus can exert its lethal effect.214 Data are lacking to evaluate the impact of adenovirus vector on apoptosis during cancer gene therapy. However, at least one salient application of one of the abovedescribed mechanisms has been tested. An E1B-55 kDadeficient adenovirus, unable to bind and block P53 replicates in, and kills, p53-deficient tumor cells but not cells with functional p53. Tested in vitro on p53-defective glioblastoma, uterine, colon and pancreas carcinoma cells, as well as in nude mice bearing human cervical carcinoma tumors, the defective virus exhibited a strong cytopathic effect specific for tumor cells, inducing complete regression of 60% of implanted tumors.215 Interesting results have also been provided by cationic liposomes (lipofection216), an attractive method since, by not requiring packaging cells, they are adapted to the transfection of highly cytotoxic DNA as ACD-inducing genes. The sequence of phenomena linked to liposomemediated delivery of DNA may influence cell viability. Indeed, recently published data indicate that lipofection specifically induces apoptosis in lymphocytes.217 These results are in agreement with our observations that many NSCLC adherent cells gained apoptotic morphology and became TUNEL-positive upon cationic-lipid delivery. Moreover, underlining its apoptotic nature, cytotoxicity was diminished by transfection of the ACD-inhibiting bclXL gene (Coll, unpublished observations).

Conclusions The above overview presents gene therapy of malignant disease in general as an attempt to induce apoptosis through genetic engineering. This focusing upon ACD is supported by the emerging in vivo and clinical data showing that the efficiency of the approach is accompanied by low general toxicity. Encouraging results obtained with the most used gene, p53, showed that the apoptotic program can be imposed in malignant cells by exogenous DNA constructs. But the variability of upstream ACD initiation also caused therapy to be targeted towards the progressively discovered final steps of apoptosis induction. This approach is based on the hypothesis that a short pathway between gene action and cell execution contributes to achieve reproducibly intense effects. As disclosed by quickly accumulating results, the strong cytocidal efficiency of bax, bclXS and caspase 1 supports this assumption. The same goal is also best attained by the association of ACD-aiming transfection with other methods of therapy, ie radiation and drugs, which are also inducers of apoptosis. Yet, performed either singly or in combination with

other methods, cancer gene therapy can reach only part of the cells forming a tumor and its metastases. Thus, a basic mechanism needed for tumor therapy is bystander apoptosis. The above data point to the counterintuitive fact that the attempt to induce ACD may be favored by a tumoral environment. While mechanisms underlying this peculiarity are far from elucidation, the trend is not totally unexpected. One could argue that, considering cell loss factor values, most cancers meet apoptosis-inducing genes with a sort of ‘spontaneous bystander killing’ around 90% of the newly produced tumor cells. Thus, the titles of two seminal papers may serve to formulate a final conclusion for this review. WT Shier asked:218 ‘Why study the mechanisms of cell death?’. The answer may reside in that, concentrating on killing the 10% surviving cells and using for this purpose the highly regulated mechanisms of active cell death, gene therapy could sustain the hope expressed by DE Fisher:29 ‘Apoptosis in cancer therapy: crossing the threshold’.

Acknowledgements ` This work was supported by the MESR (Ministere de ´ l’Enseignement Superieur et de la Recherche), the ´ ´ FNLCC (Federation Nationale de Lutte Contre le Cancer), the LNCC (Ligue Nationale de Lutte Contre le Cancer), ´ the Fondation pour la Recherche Medicale and the AFLM (Association Franc¸aise de Lutte Contre la Mucoviscidose). N Louis acknowledges financial support from the LNCC and A Negoescu from the COMARES ´ ` (Comite des Maladies Respiratoires de l’Isere). We thank Dr Ruth Griffin-Shea who kindly agreed to revise the English version of this manuscript.

References 1 Green DR, Bissonnette RP, Cotter TG. Apoptosis and cancer. In: DeVita VT, Hellman S, Rosenberg SA (eds). Important Advances in Oncology. JB Lippincott: Philadelphia, 1994, pp 37–52. 2 Kerr JFR, Winterford CM, Harmon BV. Apoptosis: its significance in cancer and cancer therapy. Cancer 1994; 73: 2013–2026. 3 Yang E, Korsmeyer S. Molecular thanatopsis: a discourse on the bcl2 family and cell death. Blood 1996; 88: 386–401. 4 Ko JK, Prives C. P53: puzzle and paradigm. Genes Dev 1996; 10: 1054–1072. 5 Brambilla E et al. Apoptosis-related factors P53, Bcl2, and Bax in neuroendocrine lung tumors. Am J Pathol 1996; 149: 1941–1952. 6 Perego P et al. Association between cisplatin resistance and mutation of p53 gene and reduced bax expression in ovarian carcinoma cell systems. Cancer Res 1996; 56: 556–562. 7 Krajewski S et al. Reduced expression of proapoptotic gene bax is associated with poor response rates to combination chemotherapy and shorter survival in women with metastatic breast adenocarcinoma. Cancer Res 1995; 55: 4471–4487. 8 Kerr JFR, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972; 26: 239–257. 9 Wyllie AH, Kerr JFR, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol 1980; 68: 251–305. 10 Steel GG. Cell loss as a factor in the growth rate of human tumors. Eur J Cancer 1967; 3: 381–387. 11 Kerr KM, Lamb D. Actual growth rate and tumour cell proliferation in human pulmonary neoplasms. Br J Cancer 1984; 50: 343–349. 12 Wyllie AH. The biology of cell death in tumors. Anticancer Res 1985; 5: 131–136. 13 Kerr JFR, Searle J. A suggested explanation for the paradoxically slow growth rate of basal-cell carcinomas that contain numerous mitotic figures. J Pathol 1972; 107: 41–44.

Apoptosis–based gene therapy of cancer M Favrot et al

14 Schatten WE. An experimental study of necrosis in tumors. Cancer Res 1962; 22: 286–290. 15 Sarraf CE, Bowen ID. Proportions of mitotic and apoptotic cells in a range of untreated experimental tumors. Cell Tiss Kinet 1988; 21: 45–49. 16 Staunton MJ, Gaffney EF. Tumor type is a determinant of susceptibility to apoptosis. Am J Clin Pathol 1995; 103: 300–307. 17 Bursch W et al. Determination of the length of the histological stages of apoptosis in normal liver and in altered hepatic foci of rats. Carcinogenesis 1990; 11: 847–853. 18 Arends MJ, McGregor AH, Wyllie AH. Apoptosis is inversely related to necrosis and determines net growth in tumors bearing constitutively expressed myc, ras and HPV oncogenes. Am J Pathol 1994; 144: 1045–1057. 19 Raff MC. Social control on cell survival and cell death. Nature 1992; 356: 397–399. 20 de Pinho RA, Schreiber-Agus N, Alt FW. Myc family oncogenes in the development of normal and neoplastic cells. Adv Cancer Res 1991; 57: 1–46. 21 Fotedar R, Diedrich L, Fotedar A. Apoptosis and the cell cycle. Prog Cell Cycle Res 1996; 2: 147–163. 22 Evan GI et al. Induction of apoptosis in fibroblasts by c-Myc protein. Cell 1992; 69: 119–128. 23 Dou QP, An B, Will PL. Induction of a rb phosphatase activity by anticancer drugs accompanies p53-independent G1 arrest and apoptosis. Proc Natl Acad Sci USA 1995; 92: 9019–9023. 24 Haas-Kogan DA et al. Inhibition of apoptosis by the rb gene product. EMBO J 1995; 14: 461–472. 25 Hannun YA. Apoptosis and the dilemma of cancer chemotherapy. Blood 1997; 89: 1845–1853. 26 Lennon SV, Martin SJ, Cotter TG. Dose-dependent induction of apoptosis in human tumour cell lines by widely diverging stimuli. Cell Prolif 1991; 24: 203–214. 27 McDonnell TJ, Meyn RE, Robertson LE. Implications of apoptotic cell death regulation in cancer therapy. Semin Cancer Biol 1995; 6: 53–60. 28 Tenniswood MP et al. Active cell death in hormone-dependent tissues. Cancer Metast Rev 1992; 11: 197–220. 29 Fisher DE. Apoptosis in cancer therapy: crossing the threshold. Cell 1994; 78: 539–542. 30 Roth JA, Cristiano RJ. Gene therapy for cancer: what have we done and where are we going? J Natl Cancer Inst 1997; 89: 21–39. 31 Sikora K. Gene therapy for cancer. Eur J Cancer 1991; 27: 1069–1070. 32 Weinberg RA. Tumor suppressor genes. Science 1991; 254: 1138–1146. 33 Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programed cell death. Cell 1993; 74: 609–619. 34 Tsujimoto Y et al. The t(14;18) chromosome translocations involved in B cell neoplasms result from mistakes in VDJ joining. Science 1985; 229: 1390–1393. 35 Hockenbery DM. Bcl2 in cancer, development and apoptosis. J Cell Sci 1994; 18 (Suppl.): 51–55. 36 Sedlak TW et al. Multiple Bcl2 family members demonstrate selective dimerizations with Bax. Proc Natl Acad Sci USA 1995; 92: 7834–7838. 37 Kernohan NM, Cox LS. Regulation of apoptosis by Bcl2 and its related proteins: immunocytochemical challenges and therapeutic implications. J Pathol 1996; 179: 1–3. 38 Saegusa M, Kamata Y, Isono M, Okayasu I. Bcl2 expression is correlated with a low apoptotic index and associated with progesterone receptor immunoreactivity in endometrial carcinomas. J Pathol 1996; 180: 275–282. 39 Koshida Y, Saegusa M, Okayasu I. Apoptosis, cell proliferation and expression of Bcl2 and Bax in gastric carcinomas: immunohistochemical and clinicopathological study. Br J Cancer 1997; 75: 367–373. 40 Rampino N et al. Somatic frameshift mutations in the bax gene in colon cancers of the microsatellite mutator phenotype. Science 1997; 275: 967–969.

41 Boise LH et al. BclX, a bcl2-related gene that functions as a dominant regulator of apoptotic cell death. Cell 1993; 74: 597–608. 42 Minn AJ, Boise LH, Thompson CB. BclX S antagonizes the protective effects of BclX L. J Biol Chem 1996; 271: 6306–6312. 43 Martin SJ, Green DR. Protease activation during apoptosis: death by a thousand cuts? Cell 1995; 82: 349–352. 44 Alnemri ES et al. Human ICE/CED3 protease nomenclature. Cell 1996; 87: 171. 45 Wyllie AH. Death gets a brake. Nature 1994; 369: 272–273. 46 Chinnaiyan AM et al. Molecular ordering of the cell death pathway: Bcl2 and BclX L function upstream of the CED3-like apoptotic proteases. J Biol Chem 1996; 271: 4573–4576. 47 Simonian PL, Grillot DAM, Merino R, Nunez G. Bax can antagonize BclX L during etoposide and cisplatin-induced cell death independently of its heterodimerization with BclX L. J Biol Chem 1996; 271: 22764–22772. 48 Simonian PL et al. Bax homodimerization is not required for Bax to accelerate chemotherapy-induced cell death. J Biol Chem 1996; 271: 32073–32077. 49 Xiang J, Chao DT, Korsmeyer SJ. Bax-induced cell death may not require ICE enzyme-like proteases. Proc Natl Acad Sci USA 1996; 93: 14559–14563. 50 McCarthy NJ, Whyte MKB, Gilbert CS, Evan GE. Inhibition of CED3/ICE proteases does not prevent cell death induced by oncogenes, DNA damage or the Bcl2 homologue Bak. J Cell Biol 1997; 136: 215–227. 51 Kinzler KW, Vogelstein B. Life (and death) in a malignant tumor. Nature 1996; 379: 19–20. 52 Clurman B, Groudine M. Killer in search of a motive? Nature 1997; 389: 122–123. 53 Lane DP. P53, guardian of the genome. Nature 1992; 358: 15–16. 54 Miyashita T, Harigai M, Hanada M, Reed JC. Identification of a p53 dependent negative response element in the bcl2 gene. Cancer Res 1994; 54: 3131–3135. 55 Miyashita T et al. Tumor suppressor p53 is a regulator of bcl2 and bax gene expression in vitro and in vivo. Oncogene 1994; 9: 1799–1805. 56 Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 1995; 80: 293–299. 57 Yin C, Knudson CM, Korsmeyer SJ, van Dyke T. Bax suppresses tumorigenesis and stimulates apoptosis in vivo. Nature 1997; 385: 637–640. 58 Lowe SW, Ruley HE, Jacks T, Housman DE. P53-dependent apoptosis modulates the cytotoxicity of anticancer agents. Cell 1993; 74: 957–967. 59 Lowe SW et al. P53 status and the efficacy of cancer therapy in vivo. Science 1994; 266: 807–810. 60 Clarke AR et al. Thymocyte apoptosis induced by P53-dependent and independent pathways. Nature 1993; 362: 849–852. 61 Clarke AR et al. P53 dependence of early apoptotic and proliferative responses within the mouse intestinal epithelium following g-irradiation. Oncogene 1994; 9: 1767–1773. 62 Datta R et al. Overexpression of BclXL by cytotoxic drug exposure confers resistance to ionizing radiation-induced internucleosomal DNA fragmentation. Cell Growth Differ 1995; 6: 363–370. 63 Lowe SW et al. P53 is required for radiation-induced apoptosis in mouse thymocytes. Nature 1993; 362: 847–848. 64 Momand J et al. The mdm2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 1992; 69: 1237–1245. 65 Oliner JD et al. Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature 1992; 358: 80–83. 66 Kondo S et al. MDM2 protein confers the resistance of a human glioblastoma cell line to cisplatin-induced apoptosis. Oncogene 1995; 10: 2001–2006. 67 Haupt Y, Barak Y, Oren M. Cell type-specific inhibition of p53mediated apoptosis by mdm2. EMBO J 1996; 15: 1596–1606. 68 Strasser A et al. S. Bcl2 and Fas/Apo1 regulate distinct pathways to lymphocyte apoptosis. EMBO J 1995; 14: 6136–6147.

735

Apoptosis–based gene therapy of cancer M Favrot et al

736

69 Nagata S. Apoptosis by death factor. Cell 1997; 88: 355–365. 70 O’Reilly LA, Huang DCS, Strasser A. The cell death inhibitor Bcl2 and its homologues influence control of cell cycle entry. EMBO J 1996; 15: 6979–6990. 71 Borner C. Diminished cell proliferation associated with the death-protective activity of Bcl2. J Biol Chem 1996; 271: 12695–12698. 72 Bacchetti S, Graham FL. Inhibition of cell proliferation by an adenovirus vector expressing the human wild-type p53 protein. Int J Oncol 1993; 3: 781–788. 73 Baker SJ et al. Suppression of human colorectal carcinoma cell growth by wild-type p53. Science 1990; 249: 912–915. 74 Casey G et al. Growth suppression of human breast cancer by the introduction of a wild-type p53 gene. Oncogene 1991; 6: 1791–1797. 75 Chen Y et al. Expression of wild-type p53 in human A673 cells suppresses tumorigenicity but not growth rate. Oncogene 1991; 6: 1799–1805. 76 Cirielli C et al. Adenovirus-mediated gene transfer of wild-type p53 results in melanoma cell apoptosis in vitro and in vivo. Int J Cancer 1995; 63: 673–679. 77 Eastham JA et al. In vivo gene therapy with p53 or p21 for prostate cancer. Cancer Res 1995; 55: 5151–5155. 78 Eicher SA et al. Evaluation of topical gene therapy for head and neck squamous cell carcinoma in an organotypic model. Clin Cancer Res 1996; 2: 1659–1664. 79 Hamada K et al. Adenovirus-mediated transfer of a wild-type p53 gene and induction of apoptosis in cervical cancer. Cancer Res 1996; 56: 3047–3054. 80 Harris CC. Structure and function of the p53 tumor suppressor gene: clues for rational cancer therapeutic strategies. J Natl Cancer Inst 1996; 88: 1442–1455. 81 Isaacs WB, Carter BS, Ewing CM. Wild-type p53 suppresses growth of human prostate cancer cells containing mutant p53 alleles. Cancer Res 1991; 51: 4716–4720. 82 Kimura M et al. Inability to induce the alteration of tumorigenicity and chemosensitivity of p53-null human pancreatic carcinoma cells after the transduction of wild-type p53 gene. Anticancer Res 1997; 17: 879–883. 83 Liu TJ et al. Growth suppression of human head and neck cancer cells by the introduction of a wild-type p53 gene via a recombinant adenovirus. Cancer Res 1994; 54: 3662–3667. 84 Liu TJ et al. Apoptosis induction mediated by wild-type p53 adenoviral gene transfer in squamous cell carcinoma of the head and neck. Cancer Res 1995; 55: 3117–3122. 85 Matozaki T et al. P53 gene mutations in human gastric cancer: wild-type p53 but not mutant p53 suppresses growth of human gastric cancer cells. Cancer Res 1992; 52: 4335–4341. 86 Ramqvist T et al. Wild-type p53 induces apoptosis in a Burkitt lymphoma line that carries mutant p53. Oncogene 1993; 8: 1495–1500. 87 Santoso JT et al. Adenovirus based p53 gene therapy in ovarian cancer. Gynecol Oncol 1995; 59: 171–178. 88 Shaw P et al. Induction of apoptosis by wild-type p53 in a human colon tumor-derived cell line. Proc Natl Acad Sci USA 1992; 89: 4495–4499. 89 Vikhanskaya F, Erba E, d’Incalci M, Broggini M. Introduction of wild-type p53 in a human ovarian cancer cell line not expressing endogenous p53. Nucleic Acids Res 1994; 22: 1012– 1017. 90 Wang J, Bucana CD, Roth JA, Zhang WW. Apoptosis induced in human osteosarcoma cells is one of the mechanisms for the cytocidal effect of Ad5CMV-p53. Cancer Gene Ther 1995; 2: 9–17. 91 Wang NP, To H, Lee WH, Lee EYHP. Tumor suppressor activity of rb and p53 genes in human breast carcinoma cells. Oncogene 1993; 8: 279–288. 92 Yang B, Stambrook PJ, Markowitz SD. Wild-type p53 demonstrates functional dominance in a human colon carcinoma cell line in which it induces reversible growth arrest. Clin Cancer Res 1996; 2: 1639–1647. 93 Yang C, Cirielli C, Capogrossi MC, Passaniti A. Adenovirus-

94 95

96

97

98

99

100

101

102 103

104

105

106

107

108

109

110

111

112

113

114

115

mediated wild-type p53 expression induces apoptosis and suppresses tumorigenesis of prostatic tumor cells. Cancer Res 1995; 55: 4210–4213. Yonish-Rouach E et al. P53-mediated cell death: relationship to cell cycle control. Mol Cell Biol 1993; 13: 1415–1423. Zhang WW et al. High-efficiency gene transfer and high-level expression of wild-type p53 in human lung cancer cells mediated by recombinant adenovirus. Cancer Gene Ther 1994; 1: 5–13. Qazilbash MH et al. Cancer gene therapy using a novel adenoassociated virus vector expressing human wild-type p53. Gene Therapy 1997; 4: 675–682. Ookawa K, Tsuchida S, Adachi JI, Yokota J. Differentiation induced by rb expression and apoptosis induced by p53 expression in an osteosarcoma cell line. Oncogene 1997; 14: 1389–1396. Sala A et al. Apoptotic response to oncogenic stimuli: co-operative and antagonistic interactions between c myb and the growth suppressor p53. Cancer Res 1996; 56: 1991–1996. Wang Y et al. Reconstitution of wild-type p53 expression triggers apoptosis in a p53-negative v-myc retrovirus-induced T cell lymphoma line. Cell Growth Diff 1993; 4: 467–473. Wang Y et al. Wild-type p53-triggered apoptosis is inhibited by bcl2 in a v-myc-induced T cell lymphoma line. Oncogene 1993; 8: 3427–3431. Mercer WE et al. Negative growth regulation in a glioblastoma tumor cell line that conditionally expresses human wild-type p53. Proc Natl Acad Sci USA 1990; 87: 6166–6170. Badie B et al. Adenovirus-mediated p53 gene delivery inhibits 9L glioma growth in rats. Neurol Res 1995; 17: 209–216. ¨ Kock H et al. Adenovirus-mediated p53 gene transfer suppresses growth of human glioblastoma cells in vitro and in vivo. Int J Cancer 1996; 67: 808–815. Spitz FR et al. In vivo adenovirus-mediated p53 tumor suppressor gene therapy for colorectal cancer. Anticancer Res 1996; 16: 3415–3422. Saito Y, Ogawa K. Wild-type p53 and c-myc co-operation in generating apoptosis of a rat hepatocellular carcinoma cell line (FAA-HTC1). Oncogene 1995; 11: 1013–1018. Cai DW et al. Stable expression of the wild-type p53 gene in human lung cancer cells after retrovirus-mediated gene transfer. Hum Gene Ther 1993; 4: 617–624. Fujiwara T et al. Induction of chemosensitivity in human lung cancer cells in vivo by adenovirus mediated-transfer of the wild-type p53 gene. Cancer Res 1994; 54: 2287–2291. Katayose D, Wersto R, Cowan KH, Seth P. Effects of a recombinant adenovirus expressing Waf1/Cip1 on cell growth, cell cycle and apoptosis. Cell Growth Diff 1995; 6: 1207–1212. Fujiwara T et al. A retroviral wild-type p53 expression vector penetrates human lung cancer spheroids and inhibits growth by inducing apoptosis. Cancer Res 1993; 53: 4129–4133. Adachi JI et al. Induction of apoptosis but not G1 arrest by expression of the wild-type p53 gene in small cell lung carcinoma. Cell Growth Diff 1996; 7: 879–886. Clayman GL et al. In vivo molecular therapy with p53 adenovirus for microscopic residual head and neck squamous carcinoma. Cancer Res 1995; 55: 1–6. Clayman GL et al. Gene therapy for head and neck cancer: comparing the tumor suppressor gene p53 and a cell cycle regulator waf1/cip1(p21). Arch Otolaryngol Head Neck Surg 1996; 122: 489–493. Fujiwara T et al. Therapeutic effect of a retroviral wild-type p53 vector in an orthotopic lung cancer model. J Natl Cancer Inst 1994; 86: 1458–1462. Lesson-Wood LA et al. Systemic gene therapy with p53 reduces growth and metastases of a malignant human breast cancer in nude mice. Hum Gene Ther 1995; 6: 395–405. Radinski R et al. Terminal differentiation and apoptosis in experimental lung metastases of human osteogenic sarcoma cells by wild-type p53. Oncogene 1994; 9: 1877–1883.

Apoptosis–based gene therapy of cancer M Favrot et al

116 Yang B, Eshelman JR, Berger NA, Markowitz SD. Wild-type p53 protein potentiates cytotoxicity of therapeutic agents in human colon cancer cells. Clin Cancer Res 1996; 2: 1649–1657. 117 Gomez-Manzano C et al. Adenovirus-mediated transfer of the p53 gene produces rapid and generalized death of human glioma cells via apoptosis. Cancer Res 1996; 56: 694–699. 118 Chen X, Ko LJ, Jayaraman L, Prives C. P53 levels, functional domains and DNA damage determine the extent of the apoptotic response of tumor cells. Genes Dev 1996; 10: 2438–2451. 119 Ronen D et al. Induction of HL60 cells to undergo apoptosis is determined by high levels of wild-type p53 protein whereas differentiation of the cells is mediated by lower p53 levels. Cell Growth Diff 1996; 7: 21–30. 120 Wang XW et al. The XPB and XPD DNA helicases are components of the p53-mediated apoptosis pathway. Genes Dev 1996; 10: 1219–1232. 121 Lassus P, Frelin M, Piette J, Hibner U. Anti-apoptotic activity of low levels of wild-type p53. EMBO J 1996; 15: 4566–4573. 122 Selivanova G et al. Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nature Med 1997; 3: 632–638. 123 Xu M et al. Parenteral gene therapy with p53 inhibits human breast tumors in vivo through a bystander mechanism without evidence of toxicity. Hum Gene Ther 1997; 8: 177–185. 124 Hamel W, Magnelli L, Chiarugi VP, Israel MA. HSV-TK/ ganciclovir-mediated apoptotic death by bystander cells. Cancer Res 1996; 56: 2696–2702. 125 Samejima Y, Meruelo D. Bystander killing induces apoptosis and is inhibited by forskolin. Gene Therapy 1995; 2: 50–58. 126 Freeman SM et al. The bystander effect: tumor regression when a fraction of the tumor mass is genetically modified. Cancer Res 1993; 53: 5274–5283. 127 Whartenby KA et al. The biology of cancer gene therapy. Lab Invest 1995; 72: 131–145. 128 Mesnil M et al. Bystander killing of cancer cells by HSV-TK is mediated by connexins. Proc Natl Acad Sci USA 1996; 93: 1831–1835. 129 Chen L, Waxman DJ. Intratumoral activation and enhanced chemotherapeutic effect of oxazaphosphorines following cytochrome P450 gene transfer: development of a combined chemotherapy/cancer gene therapy strategy. Cancer Res 1995; 55: 581–589. 130 Wu JK et al. Bystander tumoricidal effect in the treatment of experimental brain tumors. Neurosurgery 1994; 35: 1094–1103. 131 Negoescu A et al. Contribution of apoptosis to the phenotypic changes of adrenocortical cells in primary culture. Mol Cell Endocrinol 1995; 110: 175–184. 132 Barba D, Hardin J, Sadelain M, Gage FH. Development of antitumor immunity following HSV-TK-mediated killing of experimental brain tumors. Proc Natl Acad Sci USA 1994; 91: 4348– 4352. 133 Vile RG et al. Systemic gene therapy of murine melanoma using tissue specific expression of the HSV-TK gene involves an immune component. Cancer Res 1994; 54: 6228–6234. 134 Dilber MS et al. Suicide gene therapy for plasma cell tumors. Blood 1996; 88: 2192–2200. 135 Dilber MS, Smith CIE. Suicide genes and bystander killing: local and distant effects. Gene Therapy 1997; 4: 273–274. 136 Wilson KM et al. HSV-TK gene therapy in head and neck squamous cell carcinoma: enhancement by the local and distant bystander effect. Arch Otolaryngol Head Neck Surg 1996; 122: 746–749. 137 Waldman T et al. Cell-cycle arrest versus cell death in cancer therapy. Nature Med 1997; 3: 1034–1036. 138 Levine AJ. P53, the cellular gatekeeper for growth and division. Cell 1997; 88: 323–331. 139 Liebermann DA, Hoffman B, Steinman RA. Molecular controls of growth arrest and apoptosis: p53 dependent and independent pathways. Oncogene 1995; 11: 199–210. 140 Caelles C, Helmberg A, Karin M. P53-dependent apoptosis in

141 142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161 162

the absence of transcriptional activation of P53-target genes. Nature 1994; 370: 220–223. Pietenpol JA, Vogelstein B. No room at the p53 inn. Nature 1993; 365: 17–18. Owen-Schaub LB et al. Wild-type human p53 and a temperature-sensitive mutant induce Fas/APO1 expression. Mol Cell Biol 1995; 15: 3032–3040. Tamura T et al. Induction of Fas-mediated apoptosis in p53transfected human colon carcinoma cells. Oncogene 1995; 11: 1939–1946. van Meir EG et al. Release of an inhibitor of angiogenesis upon induction of wild-type p53 expression in glioblastoma cells. Nature Genet 1994; 8: 171–176. Dameron KM et al. Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin1. Science 1994; 265: 1582– 1584. Kieser A et al. Mutant p53 potentiates protein kinase C induction of vascular endothelial growth factor expression. Oncogene 1994; 9: 963–969. Roth JA. Clinical protocol: modification of tumor suppressor gene expression in non-small cell lung cancer (NSCLC) with a retroviral vector expressing wild-type (normal) p53. Hum Gene Ther 1996; 7: 861–874. Roth JA et al. Retrovirus mediated wild-type p53 gene transfer to tumors of patients with lung cancer. Nature Med 1996; 2: 985–991. Chase GA et al. Department of Health and Human Services National Institutes of Health Recombinant DNA Advisory Committee minutes of meeting: 4–5 December 1995. Hum Gene Ther 1997; 8: 1137–1186. O’Connor PM et al. Role of the p53 tumor suppressor gene in cell cycle arrest and radiosensitivity of Burkitt’s lymphoma cell lines. Cancer Res 1993; 53: 4776–4780. Gjerset RA et al. Use of wild-type p53 to achieve complete treatment sensitization of tumor cells expressing endogenous mutant p53. Molec Carcinog 1995; 14: 275–285. Spitz FR et al. Adenoviral-mediated wild-type p53 gene expression sensitizes colorectal cancer cells to ionizing radiation. Clin Cancer Res 1996; 2: 1665–1671. Xu GW et al. Tissue-specific growth suppression and chemosensitivity promotion in human hepatocellular carcinoma cells by retroviral-mediated transfer of the wild-type p53 gene. Hepatology 1996; 24: 1264–1268. Nguyen DM et al. Disruption of p53 function sensitizes breast cancer MCF7 cells to cisplatin and pentoxifylline. Cancer Res 1995; 55: 1649–1654. Wahl AF et al. Loss of normal p53 function confers sensitization to Taxol by increasing G2/M arrest and apoptosis. Nature Med 1996; 2: 72–79. Fan S et al. Disruption of p53 function sensitizes breast cancer MCF7 cells to cisplatin and pentoxifylline. Cancer Res 1995; 55: 1649–1654. Goldsmith ME et al. Wild-type p53 stimulates expression from the human multidrug resistance promoter in a p53-negative cell line. J Biol Chem 1995; 270: 1894–1898. Roth JA. Clinical protocol: modification of tumor suppressor gene expression and induction of apoptosis in non-small cell lung cancer (NSCLC) with an adenovirus vector expressing wild-type p53 and cisplatin. Hum Gene Ther 1996; 7: 1013–1030. Bayever E et al. Selective cytotoxicity to human leukemic myeloblasts produced by oligodeoxyribonucleotide phosphorothioates complementary to p53 nucleotide sequences. Leuk Lymphoma 1994; 12: 223–231. Bishop MR et al. Phase I trial of and antisense oligonucleotide OL(1) p53 in hematologic malignancies. J Clin Oncol 1996; 14: 1320–1326. El-Deiry WS et al. Waf1/cip1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res 1994; 54: 1169–1174. Harper JW et al. The p21 cdk-interacting protein cip1 is a potent inhibitor of cyclin-dependent kinases. Cell 1993; 75: 805–816.

737

Apoptosis–based gene therapy of cancer M Favrot et al

738

163 Xiong Y et al. P21 is a universal inhibitor of cyclin kinases. Nature 1993; 366: 701–704. 164 Hooper ML. The role of p53 and rb1 genes in cancer, development and apoptosis. J Cell Sci 1994; 18: 13–17. 165 White E. P53, guardian of Rb. Nature 1994; 371: 21–22. 166 Jung JM et al. Inhibition of human glioblastoma cell growth by Waf1/Cip1 can be attenuated by mutant P53. Cell Growth Diff 1995; 6: 909–913. 167 Sheikh MS, Rochefort H, Garcia M. Overexpression of p21/waf1/cip1 induces growth arrest, giant cell formation and apoptosis in human breast carcinoma cell lines. Oncogene 1995; 11: 1899–1905. 168 Skotzko M et al. Retroviral vector-mediated gene transfer of antisense cyclin G1 (CYCG1) inhibits proliferation of human osteogenic sarcoma cells. Cancer Res 1995; 55: 5493–5498. 169 Yang ZY et al. The P21/Waf1 cyclin-dependent kinase inhibitor suppresses tumorigenicity in vivo. Nature Med 1995; 1: 1052– 1056. 170 Jin X et al. Cell cycle arrest and inhibition of tumor cell proliferation by the p16 INK4 gene mediated by adenovirus vector. Cancer Res 1995; 55: 3250–3253. 171 Sandig V et al. Adenovirally transfected p16 INK4/CDKN2 and p53 genes co-operate to induce apoptotic tumor cell death. Nature Med 1997; 3: 313–319. 172 Clarke MF et al. A recombinant bclXS adenovirus selectively induces apoptosis in cancer cells but not in normal bone marrow cells. Proc Natl Acad Sci USA 1995; 92: 11024–11028. 173 Dole MG et al. BclXS enhances adenoviral vector-induced apoptosis in neuroblastoma cells. Cancer Res 1996; 56: 5734– 5740. 174 Ealovega MW et al. BclXS gene therapy induces apoptosis of human mammary tumors in nude mice. Cancer Res 1996; 56: 1965–1969. 175 Sumantran VN et al. Overexpression of bclX S sensitizes MCF7 cells to chemotherapy-induced apoptosis. Cancer Res 1995; 55: 2507–2510. 176 Sakakura C et al. Overexpression of bax sensitizes human breast cancer MCF7 cells to radiation-induced apoptosis. Int J Cancer 1996; 67: 101–105. 177 Strobel T, Swanson L, Korsmeyer S, Cannistra SA. Bax enhances paclitaxel-induced apoptosis through a P53-independent pathway. Proc Natl Acad Sci USA 1996; 93: 14094– 14099. 178 Coll J-L et al. Antitumor activity of Bax and P53 naked gene transfer in lung cancer. In vitro and in vivo analysis. Hum Gene Ther (submitted). 179 Yu JS et al. Retroviral delivery and tetracycline-dependent expression of IL-1b-converting enzyme (ICE) in a rat glioma model provides controlled induction of apoptotic death in tumor cells. Cancer Res 1996; 56: 5423–5427. 180 Weller M et al. Fas/APO1 gene transfer for human malignant glioma. Cancer Res 1995; 55: 2936–2944. 181 Asher AL et al. Murine tumor cells transduced with the gene for TNF a. J Immunol 1991; 146: 3227–3234. 182 Hwu P et al. Functional and molecular characterization of tumor-infiltrating lymphocytes transduced with TNF␣ cDNA for the gene therapy of cancer in humans. J Immunol 1993; 150: 4104–4115. 183 Tos AG, Cignetti A, Rovera G, Foa R. Retroviral vectormediated transfer of the TNF␣ gene into human cancer cells restores an apoptotic cell death program and induces a bystander killing effect. Blood 1996; 87: 2486–2495. 184 Feng M et al. Neoplastic reversion accomplished by high efficiency adenoviral-mediated delivery of an anti-ras ribozyme. Cancer Res 1995; 55: 2024–2028. 185 Georges RN et al. Prevention of orthotopic human lung cancer growth by intratracheal instillation of a retroviral antisense Kras construct. Cancer Res 1993; 53: 1743–1746. 186 Mukhopadhyay T, Tainski M, Cavender AC, Roth JA. Specific inhibition of K ras expression and tumorigenicity of lung cancer cells by antisense DNA. Cancer Res 1991; 51: 1744–1748.

187 Zhang Y et al. Retroviral vector-mediated transduction of Kras antisense RNA into human lung cancer cells inhibits expression of the malignant phenotype. Hum Gene Ther 1993; 4: 451–460. 188 Kasid U et al. Effect of antisense c-raf1 on tumorigenicity and radiation sensitivity of a human squamous carcinoma. Science 1989; 243: 1354–1356. 189 Ledwith B et al. Antisense fos RNA causes partial reversion of the transformed phenotypes induced by the c Ha ras oncogene. Mol Cell Biol 1990; 10: 1545–1555. 190 Sklar MD et al. Depletion of c-myc with specific antisense sequences reverses the transformed phenotype in ras oncogene-transformed NIH3T3 cells. Mol Cell Biol 1991; 11: 3699– 3710. 191 Wang HG et al. Apotosis regulation by interaction of Bcl2 protein and Raf 1 kinase. Oncogene 1994; 9: 2751–2756. 192 Nooter K et al. Constitutive expression of the c-H-ras oncogene inhibits doxorubicin-induced apoptosis and promotes cell survival in a rhabdomyosarcoma cell line. Br J Cancer 1995; 71: 556–561. 193 Bedi A et al. Inhibition of apoptosis by Bcr-Abl in chronic myeloid leukemia. Blood 1994; 83: 2038–2044. 194 McGahon A et al. Bcr-abl maintains resistance of chronic myelogenous leukemia cells to apoptotic cell death. Blood 1994; 5: 1179–1187. 195 Smetsers TFCM et al. Antisense bcr-abl oligonucleotides induce apoptosis in the Philadelphia chromosome-positive cell line BV173. Leukemia 1994; 8: 129–140. 196 Skorski T et al. Antisense oligodeoxynucleotide combination therapy of primary chronic myelogenous leukemia blast crisis in SCID mice. Blood 1996; 88: 1005–1012. 197 Skorski T et al. Treatment of Philadelphia leukemia in severe combined immunodeficient mice by combination of cyclophosphamide and bcr-abl antisense oligodeoxynucleotides. J Natl Cancer Inst 1997; 89: 124–133. 198 Keith FJ, Bradbury DA, Zhu YM, Russell NH. Inhibition of bcl2 with antisense oligonucleotides induces apoptosis and increases the sensitivity of AML blasts to Ara-C. Leukemia 1995; 9: 131–138. 199 Kitada S et al. Reversal of chemoresistance of lymphoma cells by antisense-mediated reduction of bcl2 gene expression. Antisense Res Dev 1994; 4: 71–79. 200 Reed JC et al. Antisense mediated inhibition of bcl2 proto-oncogene expression and leukemic cell growth and survival: comparisons of phosphodiester and phosphorothioate oligodeoxynucleotides. Cancer Res 1990; 50: 6565–6570. 201 Reed JC et al. Bcl2 mediated tumorigenicity of a human T lymphoyd cell line: synergy with myc and inhibition by bcl2 antisense. Proc Natl Acad Sci USA 1990; 87: 3660–3664. 202 Cotter FE et al. Antisense oligonucleotides suppress B cell lymphoma growth in a SCID-hu mouse model. Oncogene 1994; 9: 3049–3055. 203 Webb A et al. Bcl2 antisense therapy in patients with nonHodgkin lymphoma. Lancet 1997; 349: 1137–1141. 204 Kondo S et al. Bcl2 gene enables rescue from in vitro myelosuppression (bone marrow cell death) induced by chemotherapy. Br J Cancer 1994; 70: 421–426. 205 Dole MG et al. BclXL is expressed in neuroblastoma cells and modulates chemotherapy-induced apoptosis. Cancer Res 1995; 55: 2576–2582. 206 Minn AJ, Rudin CM, Boise LH, Thompson CB. Expression of bclX L can confer a multidrug resistance phenotype. Blood 1995; 86: 1903–1910. 207 Debbas M, White E. Wild-type p53 mediates apoptosis by E1A, which is inhibited by E1B. Genes Dev 1993; 7: 546–554. 208 Lowe SW, Ruley HE. Stabilization of the p53 tumor suppressor is induced by adenovirus 5 E1A and accompanies apoptosis. Genes Dev 1993; 7: 535–545. 209 Teodoro JC, Shore GC, Branton PE. Adenovirus E1A proteins induce apoptosis by both p53-dependent and p53-independent mechanisms. Oncogene 1995; 11: 467–474.

Apoptosis–based gene therapy of cancer M Favrot et al

210 Boyd JM et al. Adenovirus E1B 19 kDa and Bcl2 proteins interact with a common set of cellular proteins. Cell 1994; 79: 341–351. 211 Han J et al. The E1B protein blocks apoptosis by interacting with and inhibiting the p53-inducible and death-promoting Bax protein. Genes Dev 1996; 10: 461–477. 212 Rao L et al. The adenovirus E1A proteins induce apoptosis, which is inhibited by the E1B 19 kDa and Bcl2 proteins. Proc Natl Acad Sci USA 1992; 89: 7742–7746. 213 White E, Cipriani R, Sabbatini P, Denton A. Adenovirus E1B 19 kDa protein overcomes the cytotoxicity of E1A proteins. J Virol 1991; 65: 2968–2978.

214 Brough DE et al. A gene transfer vector-cell line system for complete functional complementation of adenovirus early regions E1 and E4. J Virol 1996; 70: 6497–6501. 215 Bischoff JR et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 1996; 274: 373–376. 216 Felgner PL et al. Nomenclature for synthetic gene delivery systems. Hum Gene Ther 1997; 8: 511–512. 217 Ebert O et al. Lymphocyte apoptosis: induction by gene transfer techniques. Gene Therapy 1997; 4: 296–302. 218 Shier WT. Why study mechanisms of cell death? Meth Achiev Exp Pathol 1988; 13: 1–17.

739