Vol. 7, 2955–2957, October 2001 Clinical Cancer Research 2955

Editorial Where to Next with for Cancer Therapy?

Malcolm A. Smith1 and Barry Anderson 13-cis-RA, and 9-cis-RA causes decreased tumor cell prolifer- Pediatric Section, Cancer Therapy Evaluation Program, National ation and morphological differentiation. A Phase 2 trial of single Cancer Institute, Bethesda, Maryland 20892 agent 13-cis-RA demonstrated minimal antitumor response in children with recurrent or progressive neuroblastoma receiving a dose of 100 mg/m2/day (10). However, complete responses The report by Adamson and colleagues describes a Phase 1 were seen among children with residual tumor in the bone trial in children of 9-cis-RA2 (1). This report follows by nearly marrow who were treated in a Phase 1 trial evaluating higher 10 years the report of the pediatric Phase 1 evaluation of ATRA doses of 13-cis-RA given in two-week pulses (maximum toler- and by more than a decade the initial reports that ATRA induced ated dose, 160 mg/m2/day; Ref. 11). Subsequently, improved high remission rates in patients with APL. The authors conclude event-free survival for children with advanced stage neuroblas- their report by noting the continuing challenge of identifying the toma was demonstrated in a Phase 3 study of patients random- optimal and the optimal way to integrate retinoids into ized after completion of cytotoxic therapy to receive 6 months multiagent treatment regimens. In addressing this challenge, we 2 first briefly summarize those malignancies for which retinoids of maintenance treatment using 13-cis-RA (160 mg/m /day) on have definitively shown therapeutic benefit, and then we discuss the 2-week pulsed schedule (9). The clinical importance of clinical research opportunities for retinoids and how these can 13-cis-RA dosage for antitumor effectiveness has been sug- be prioritized. gested by the lack of 13-cis-RA benefit among children with Three retinoids have proven oncological utility: ATRA, a advanced neuroblastoma randomized to receive a lower daily ϳ 2 naturally occurring retinoid that binds to and activates RAR ␣, dose (0.75 mg/kg/day or 30 mg/m /day) after high-dose chem- ␤, and ␥; 13-cis-RA which likely acts as an ATRA prodrug; and otherapy and autologous stem cell transplant (12, 13). (Targretin, LGD1069), which selectively binds and What criteria ought to be used in selecting among these and activates RXR␣, ␤, and ␥. Best defined among the oncological other retinoids for future clinical evaluation? To the extent that indications of retinoids is the use of ATRA for APL. The basis the primary goal of treatment is cure and not palliation, an for the dramatic efficacy of ATRA against APL is the ability of important criterion is the ability of the agent to kill tumor cells pharmacological doses of ATRA to overcome the repression of or to enhance tumor cell killing when combined with other signaling caused by the PML-RAR␣ fusion protein at physio- anticancer drugs. Given this criterion, a group of retinoids of logical ATRA concentrations. Restoration of signaling leads to particular interest are the apoptogenic retinoids, as exemplified differentiation of APL cells and then to postmaturation apopto- by fenretinide [N-(4-hydroxyphenyl) retinamide] and CD437/6- sis (2). A series of randomized clinical trials have defined the [3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalenecarboxylic acid. benefit for combining ATRA with chemotherapy during induc- Fenretinide at micromolar concentrations is able to induce tion therapy and also the utility of using ATRA as maintenance apoptosis in a number of different types of cancer cells, includ- therapy (3–5). ing neuroblastoma (14, 15), non-small cell lung cancer (16), Bexarotene is approved by the FDA for the treatment of cutaneous squamous cell carcinoma (17), and others. Although ␥ cutaneous manifestations of cutaneous T-cell lymphoma in pa- fenretinide appears to be a potent transactivator with RAR and ␤ ␣ ␣ tients whose disease is refractory to at least one prior systemic a moderate activator with RAR (but not RAR and RXR ; therapy (6). Consistent with the RXR specificity of bexarotene, Ref. 18) and can induce RAR-dependent differentiation and ATRA toxicities such as cheilitis and headache appear to be less apoptosis (19, 20), it can also induce apoptosis through RAR- prominent in patients receiving bexarotene (7, 8). independent mechanisms (16, 19, 21). For some cancer cells, the 13-cis-RA improves outcome for children with advanced production of ROS plays a role in fenretinide-induced apoptosis stage neuroblastoma when given at high doses after intensive (16, 17). Retinoids such as ATRA, 13-cis-RA, and 9-cis-RA do consolidation chemotherapy (9). For many years, it has been not induce ROS (15, 22, 23). The induction of ROS is a rapid known that exposure of neuroblastoma cell lines to ATRA, cellular response to fenretinide exposure (17, 20, 23) and is not dependent on RAR activation (20, 22). Induction of ceramide synthesis may also play a role in fenretinide-induced apoptosis (14, 24). The initial clinical trials of fenretinide used relatively low doses (200 mg/day) that achieved serum levels of approx- Received 6/27/01; accepted 7/03/01. imately 1 ␮M (25). More recently, Phase 1 studies of fenretinide The costs of publication of this article were defrayed in part by the have demonstrated that much higher doses of fenretinide are payment of page charges. This article must therefore be hereby marked 2 tolerated (Ͼ500 mg/m /day) and that the 5–10-␮M concentra- advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. tions required to induce apoptosis in vitro can be achieved in at 1 To whom requests for reprints should be addressed, at Pediatric least some pediatric patients (26). Section, CTEP, Room 7025, EPN, 6130 Executive Boulevard, Bethesda, CD437, like fenretinide, induces apoptosis in a variety of MD 20892. Phone: (301) 496-2522; E-mail: [email protected]. 2 cancer cell types. CD437 selectively binds to and transactivates The abbreviations used are: 9-cis-RA, 9-cis-; ATRA, ␥ all-trans retinoic acid; APL, acute promyelocytic leukemia; RAR, reti- RAR , but is able to induce apoptosis by mechanisms independ- noic acid receptor; 13-cis-RA, 13-cis-retinoic acid; RXR, retinoid X ent of RAR transcriptional activation (27, 28). In lung and receptor; ROS, reactive oxygen species; HDAC, histone deacetylase. prostate cancer cell lines, an apoptosis-inducing CD437 ana- Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2001 American Association for Cancer Research. 2956 Editorial

logue induces expression and mitochondrial translocation of often, preclinical models provide insufficient insight into the transcription factor TR3/Nur77 and induces inner mitochondrial best use of an agent to achieve clinical benefit. Nonetheless, membrane depolarization and mitochondrial release of cyto- decisions concerning prioritizing clinical research opportunities chrome c (29, 30). CD437 can also up-regulate expression of a must be made based on the best available information, however number of apoptosis-related genes, including Killer/DR5, Bax, imperfect that information is. To the extent possible, it will be and Fas (28, 31, 32). This interesting compound has not been important to determine whether the signaling pathways identi- evaluated in the clinical setting. fied as important in preclinical models are indeed modulated in Concerning the use of retinoids in multiagent combina- the clinical setting. This closing of the loop between preclinical tions, of particular interest is the use of retinoids in combination and clinical observations allows confirmation (or refutation) of with HDAC inhibitors. The biological basis for this combination hypotheses concerning critical factors associated with retinoid is the ability of non-liganded RARs to recruit HDAC complexes antitumor activity, thereby permitting more rational applications leading to transcriptional repression of target genes (33). For of retinoids in oncological practice. APL, resistance to ATRA can in some cases be overcome by combination therapy using ATRA with a HDAC inhibitor (34, REFERENCES 35). For some non-promyelocytic cases of acute myelogenous leukemia, the poor sensitivity to retinoid-induced differentiation 1. Adamson, P. C., Widemann, B. C., Reaman, G. H., Seibel, N. L., Murphy, R. F., Gillespie, A. F., and Balis, F. M. A Phase I trial and may reflect HDAC-dependent repression of retinoid signaling pharmacokinetic study of 9-cis-retinoic acid (ALRT1057) in pediatric pa- pathways, and HDAC inhibition can restore ATRA-induced tients with refractory cancer. A joint Pediatric Oncology Branch. NCI and differentiation (36). For neuroblastoma, combination treatment Children’s Cancer Group Study. Clin. Cancer Res., 7: 3034–3039, 2001. (either in vitro or in vivo) of neuroblastoma cells with ATRA 2. Altucci, L., Rossin, A., Raffelsberger, W., Reitmair, A., Chomienne, and a HDAC inhibitor leads to greater antitumor activity than C., and Gronemeyer, H. Retinoic acid-induced apoptosis in leukemia cells is mediated by paracrine action of tumor-selective death ligand that obtained with either single agent (37, 38). Clinical evalua- TRAIL. Nat. Med., 7: 680–686, 2001. tions of retinoids with HDAC inhibitors have to date been 3. Fenaux, P., Chastang, C., Chevret, S., Sanz, M., Dombret, H., limited to the combination of ATRA with phenylbutyrate (35), Archimbaud, E., Fey, M., Rayon, C., Huguet, F., Sotto, J. J., Gardin, C., but this strategy warrants continued consideration given the Makhoul, P. C., Travade, P., Solary, E., Fegueux, N., Bordessoule, D., availability of new HDAC inhibitors in clinical evaluation [e.g., Miguel, J. S., Link, H., Desablens, B., Stamatoullas, A., Deconinck, E., MS-275, suberoylanilide hydroxamic acid (SAHA), and CI- Maloisel, F., Castaigne, S., Preudhomme, C., and Degos, L. A random- ized comparison of all-trans-retinoic acid (ATRA) followed by chem- 994]. otherapy and ATRA plus chemotherapy and the role of maintenance The effective use of ATRA with conventional cytotoxic therapy in newly diagnosed acute promyelocytic leukemia. The Euro- agents for APL (see above) provides proof-of-principle for the pean APL Group. Blood, 94: 1192–1200, 1999. potential utility of combining retinoids with cytotoxic agents. 4. Fenaux, P., Chevret, S., Guerci, A., Fegueux, N., Dombret, H., Combinations of retinoids with conventional cytotoxic agents Thomas, X., Sanz, M., Link, H., Maloisel, F., Gardin, C., Bordessoule, D., Stoppa, A. M., Sadoun, A., Muus, P., Wandt, H., Mineur, P., have shown promise in some preclinical models, including Whittaker, J. A., Fey, M., Daniel, M. T., Castaigne, S., and Degos, L. combinations of 9-cis-RA with cisplatin (39) and combinations Long-term follow-up confirms the benefit of all-trans retinoic acid in of fenretinide with either cisplatin or etoposide (15, 40, 41). The acute promyelocytic leukemia. European APL group. Leukemia (Bal- supra-additive activity of fenretinide in combination with cyto- timore), 14: 1371–1377, 2000. toxic agents appears to be associated with the ability of fen- 5. Tallman, M. S., Andersen, J. W., Schiffer, C. A., Appelbaum, F. R., retinide to induce ROS (15). Feusner, J. H., Ogden, A., Shepherd, L., Willman, C., Bloomfield, C. D., Rowe, J. M., and Wiernik, P. H. All-trans-retinoic acid in acute promyelo- Other retinoid combinations of potential utility include cytic leukemia [see comments; published erratum appears in N. Engl. retinoids plus receptor tyrosine kinase inhibitors (42) and com- J. Med., 337: 1639, 1997]. N. Engl. J. Med., 337: 1021–1028, 1997. binations of fenretinide with modulators of ceramide metabo- 6. Khuri, F. R., Rigas, J. R., Figlin, R. A., Gralla, R. J., Shin, D. M., lism (24). The ability of CD437 to up-regulate DR4 and DR5 Munden, R., Fox, N., Huyghe, M. R., Kean, Y., Reich, S. D., and Hong, death receptor expression in some cancer cell lines suggests the W. K. Multi-institutional Phase I/II trial of oral bexarotene in combi- nation with cisplatin and vinorelbine in previously untreated patients possible utility for the combination of CD437 with TRAIL, with advanced non-small cell lung cancer. J. Clin. Oncol., 19: 2626– should these compounds enter clinical evaluation (32). 2637, 2001. There are no clear roadmaps for prioritizing which retinoid 7. Miller, V. A., Benedetti, F. M., Rigas, J. R., Verret, A. L., Pfister, D. G., or combination strategy should be put forward for clinical Straus, D., Kris, M. G., Crisp, M., Heyman, R., Loewen, G. R., Truglia, evaluation. Possible characteristics arguing for prioritization of J. A., and Warrell, R. P., Jr. Initial clinical trial of a selective retinoid X a retinoid, alone or in combination, include: (a) the biological receptor ligand, LGD1069. J. Clin. Oncol., 15: 790–795, 1997. plausibility for anticancer activity based on retinoid-induced 8. Rizvi, N. A., Marshall, J. L., Dahut, W., Ness, E., Truglia, J. A., Loewen, G., Gill, G. M., Ulm, E. H., Geiser, R., Jaunakais, D., and modifications of critical survival and apoptosis signaling path- Hawkins, M. J. A Phase I study of LGD1069 in adults with advanced ways for the cancer under study; and (b) the reproducibility of cancer. Clin. Cancer Res., 5: 1658–1664, 1999. in vitro and in vivo antitumor activity in multiple cell lines 9. Matthay, K. K., Villablanca, J. G., Seeger, R. C., Stram, D. O., (xenografts) at concentrations of agent(s) that are achievable in Harris, R. E., Ramsay, N. K., Swift, P., Shimada, H., Black, C. T., patients. As an example of the application of the latter criterion, Brodeur, G. M., Gerbing, R. B., and Reynolds, C. P. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, the data reported by Adamson argue against prioritizing 9-cis- autologous bone marrow transplantation, and 13-cis-retinoic acid. Chil- RA for neuroblastoma (1), because the 9-cis-RA serum levels dren’s Cancer Group. N. Engl. J. Med., 341: 1165–1173, 1999. achievable in young children are well below the 1-␮M levels 10. Finklestein, J. Z., Krailo, M. D., Lenarsky, C., Ladisch, S., Blair, G. K., associated with growth inhibition in preclinical studies (43). Too Reynolds, C. P., Sitarz, A. L., and Hammond, G. D. 13-cis-retinoic acid

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2001 American Association for Cancer Research. Clinical Cancer Research 2957

(NSC 122758) in the treatment of children with metastatic neuroblastoma the synthetic retinoid CD437 in human lung cancer cells. Cancer Res., unresponsive to conventional chemotherapy: report from the Childrens 60: 6537–6543, 2000. Cancer Study Group. Med. Pediatr. Oncol, 20: 307–311, 1992. 29. Li, H., Kolluri, S. K., Gu, J., Dawson, M. I., Cao, X., Hobbs, P. D., 11. Villablanca, J. G., Khan, A. A., Avramis, V. I., Seeger, R. C., Lin, B., Chen, G., Lu, J., Lin, F., Xie, Z., Fontana, J. A., Reed, J. C., and Matthay, K. K., Ramsay, N. K., and Reynolds, C. P. Phase I trial of Zhang, X. Cytochrome c release and apoptosis induced by mitochon- 13-cis-retinoic acid in children with neuroblastoma following bone drial targeting of nuclear orphan receptor TR3. Science (Wash. DC), marrow transplantation. J. Clin. Oncol., 13: 894–901, 1995. 289: 1159–1164, 2000. 12. Kohler, J. A., Imeson, J., Ellershaw, C., and Lie, S. O. A random- 30. Dawson, M. I., Hobbs, P. D., Peterson, V. J., Leid, M., Lange, ized trial of 13-Cis retinoic acid in children with advanced neuroblas- C. W., Feng, K. C., Chen, G., Gu, J., Li, H., Kolluri, S. K., Zhang, X., toma after high-dose therapy. Br. J. Cancer, 83: 1124–1127, 2000. Zhang, Y., and Fontana, J. A. Apoptosis induction in cancer cells by a novel analogue of 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalen- 13. Matthay, K. K., and Reynolds, C. P. Is there a role for retinoids to ecarboxylic acid lacking transcriptional activation ac- treat minimal residual disease in neuroblastoma? Br. J. Cancer, 83: tivity. Cancer Res., 61: 4723–4730, 2001. 1121–1123, 2000. 31. Sun, S. Y., Yue, P., Mao, L., Dawson, M. I., Shroot, B., Lamph, 14. Maurer, B. J., Metelitsa, L. S., Seeger, R. C., Cabot, M. C., and W. W., Heyman, R. A., Chandraratna, R. A., Shudo, K., Hong, W. K., Reynolds, C. P. Increase of ceramide and induction of mixed apoptosis/ and Lotan, R. Identification of receptor-selective retinoids that are necrosis by N-(4-hydroxyphenyl)retinamide in neuroblastoma cell lines potent inhibitors of the growth of human head and neck squamous cell [see comments]. J. Natl. Cancer Inst. (Bethesda), 91: 1138–1146, 1999. carcinoma cells. Clin. Cancer Res., 6: 1563–1573, 2000. 15. Lovat, P. E., Ranalli, M., Bernassola, F., Tilby, M., Malcolm, A. J., 32. Sun, S. Y., Yue, P., Hong, W. K., and Lotan, R. Augmentation of Pearson, A. D., Piacentini, M., Melino, G., and Redfern, C. P. Syner- tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)- gistic induction of apoptosis of neuroblastoma by fenretinide or CD437 induced apoptosis by the synthetic retinoid 6-[3-(1-adamantyl)-4- in combination with chemotherapeutic drugs. Int. J. Cancer, 88: 977– hydroxyphenyl]-2-naphthalene carboxylic acid (CD437) through up- 985, 2000. regulation of TRAIL receptors in human lung cancer cells. Cancer Res., 16. Sun, S. Y., Li, W., Yue, P., Lippman, S. M., Hong, W. K., and 60: 7149–7155, 2000. Lotan, R. Mediation of N-(4-hydoxyphenyl)retinamide-induced apopto- 33. Redner, R. L., Wang, J., and Liu, J. M. Chromatin remodeling and sis in human cancer cells by different mechanisms. Cancer Res., 59: leukemia: new therapeutic paradigms. Blood, 94: 417–428, 1999. 2493–2498, 1999. 34. Warrell, R. P., Jr., He, L. Z., Richon, V., Calleja, E., and Pandolfi, 17. Hail, N., Jr., and Lotan, R. Mitochondrial permeability transition is P. P. Therapeutic targeting of transcription in acute promyelocytic a central coordinating event in N-(4-hydroxyphenyl)retinamide-induced leukemia by use of an inhibitor of histone deacetylase [see comments]. apoptosis. Cancer Epidemiol. Biomark. Prev., 9: 1293–1301, 2000. J. Natl. Cancer Inst. (Bethesda), 90: 1621–1625, 1998. 18. Fanjul, A. N., Delia, D., Pierotti, M. A., Rideout, D., Yu, J. Q., 35. Novick, S., Camacho, L., Gallagher, R., Chanel, S., Ho, R., To- Pfahl, M., and Qiu, J. 4-Hydroxyphenyl retinamide is a highly selective lentino, T., Richon, V., Pandolfi, P. P., and Warrell, R. P. Initial clinical activator of retinoid receptors. J. Biol. Chem., 271: 22441–22446, 1996. evaluation of “transcription therapy” for cancer: all-trans retinoic acid 19. Clifford, J. L., Menter, D. G., Wang, M., Lotan, R., and Lippman, plus phenylbutyrate. Blood, 94 (Suppl. 1): 61a, 11–15, 1999. S. M. Retinoid receptor-dependent and -independent effects of N-(4- 36. Ferrara, F. F., Fazi, F., Bianchini, A., Padula, F., Gelmetti, V., hydroxyphenyl)retinamide in F9 embryonal carcinoma cells. Cancer Minucci, S., Mancini, M., Pelicci, P. G., Lo, C. F., and Nervi, C. Histone Res., 59: 14–18, 1999. deacetylase-targeted treatment restores retinoic acid signaling and dif- ferentiation in acute myeloid leukemia. Cancer Res., 61: 2–7, 2001. 20. Lovat, P. E., Ranalli, M., Annichiarrico-Petruzzelli, M., Bernassola, F., Piacentini, M., Malcolm, A. J., Pearson, A. D., Melino, G., and 37. Coffey, D. C., Kutko, M. C., Glick, R. D., Swendeman, S. L., Butler, Redfern, C. P. Effector mechanisms of fenretinide-induced apoptosis in L., Rifkind, R., Marks, P. A., Richon, V. M., and LaQuaglia, M. P. Histone neuroblastoma. Exp. Cell Res, 260: 50–60, 2000. deacetylase inhibitors and retinoic acids inhibit growth of human neuro- blastoma in vitro. Med. Pediatr. Oncol., 35: 577–581, 2000. 21. Reynolds, C. P., and Chen, P. Response of neuroblastoma cell lines to retinoic acid and 13-cis-retinoic, but not to fenretinide, is mediated by 38. Coffey, D. C., Kutko, M. C., Glick, R. D., Butler, L. M., Heller, G., retinoic acid receptors. Proc. Am. Assoc. Cancer Res., 42: 186, 2001. Rifkind, R. A., Marks, P. A., Richon, V. M., and La Quaglia, M. P. The histone deacetylase inhibitor, CBHA, inhibits growth of human neuro- 22. Oridate, N., Suzuki, S., Higuchi, M., Mitchell, M. F., Hong, W. K., blastoma xenografts in vivo, alone and synergistically with all-trans and Lotan, R. Involvement of reactive oxygen species in N-(4-hydroxy- retinoic acid. Cancer Res., 61: 3591–3594, 2001. phenyl)retinamide-induced apoptosis in cervical carcinoma cells [see 39. Shalinsky, D. R., Bischoff, E. D., Gregory, M. L., Lamph, W. W., comments]. J. Natl. Cancer Inst. (Bethesda), 89: 1191–1198, 1997. Heyman, R. A., Hayes, J. S., Thomazy, V., and Davies, P. J. Enhanced 23. Delia, D., Aiello, A., Meroni, L., Nicolini, M., Reed, J. C., and Pierotti, antitumor efficacy of cisplatin in combination with ALRT1057 (9-cis M. A. Role of antioxidants and intracellular free radicals in retinamide- retinoic acid) in human oral squamous carcinoma xenografts in nude induced cell death. Carcinogenesis (Lond.), 18: 943–948, 1997. mice. Clin. Cancer Res., 2: 511–520, 1996. 24. Maurer, B. J., Melton, L., Billups, C., Cabot, M. C., and Reynolds, 40. Formelli, F., and Cleris, L. Therapeutic effects of the combination C. P. Synergistic cytotoxicity in solid tumor cell lines between N-(4- of fenretinide and all-trans-retinoic acid and of the two retinoids with hydroxyphenyl)retinamide and modulators of ceramide metabolism. cisplatin in a human ovarian carcinoma xenograft and in a cisplatin- J. Natl. Cancer Inst. (Bethesda), 92: 1897–1909, 2000. resistant sub-line. Eur. J. Cancer, 36: 2411–2419, 2000. 25. Formelli, F., Clerici, M., Campa, T., Di Mauro, M. G., Magni, A., 41. Kalemkerian, G. P., and Ou, X. Activity of fenretinide plus che- Mascotti, G., Moglia, D., De Palo, G., Costa, A., and Veronesi, U. motherapeutic agents in small-cell lung cancer cell lines. Cancer Che- Five-year administration of fenretinide: pharmacokinetics and effects on mother. Pharmacol., 43: 145–150, 1999. plasma concentrations. J. Clin. Oncol., 11: 2036–2042, 1993. 42. Giannõ`, M., Kalac, Y., Ponzanelli, I., Rambaldi, A., Terao, M., and 26. Luksch, R., Pizzitola, M. R., Lo Piccolo, M. S., Formelli, F., Montaldo, Garattini, E. Tyrosine kinase inhibitor STI571 potentiates the pharma- P. G., De Bernardi, B., Bellani, F. F., and Garaventa, A. Phase I study of cologic activity of retinoic acid in acute promyelocytic leukemia cells: fenretinide in neuroblastoma. Clin. Cancer Res., 6: 2000. effects on the degradation of RAR␣ and PML-RAR␣. Blood, 97: 27. Sun, S. Y., Yue, P., Shroot, B., Hong, W. K., and Lotan, R. Induction 3234Ð3243, 2001. of apoptosis in human non-small cell lung carcinoma cells by the novel 43. Irving, H., Lovat, P. E., Hewson, Q. C., Malcolm, A. J., Pearson, synthetic retinoid CD437. J. Cell. Physiol., 173: 279–284, 1997. A. D., and Redfern, C. P. Retinoid-induced differentiation of neuroblas- 28. Sun, S. Y., Yue, P., Hong, W. K., and Lotan, R. Induction of Fas toma: comparison between LG69, an RXR-selective analogue and 9-cis expression and augmentation of Fas/Fas ligand-mediated apoptosis by retinoic acid. Eur. J. Cancer, 34: 111Ð117, 1998.

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2001 American Association for Cancer Research. Where to Next with Retinoids for Cancer Therapy?

Malcolm A. Smith and Barry Anderson

Clin Cancer Res 2001;7:2955-2957.

Updated version Access the most recent version of this article at: http://clincancerres.aacrjournals.org/content/7/10/2955

Cited articles This article cites 40 articles, 21 of which you can access for free at: http://clincancerres.aacrjournals.org/content/7/10/2955.full#ref-list-1

Citing articles This article has been cited by 2 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/7/10/2955.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/7/10/2955. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2001 American Association for Cancer Research.