Phase II randomized, double-blind, placebo-controlled study of whole-brain irradiation with concomitant chloroquine for brain metastases

Luis L Rojas-Puentes, Marcelino Gonzalez-Pinedo, Alejando Crismatt, Alette Ortega-Gomez, Carlos Gamboa-Vignolle, Rodrigo Nuñez-Gomez, Yusmiren Dorantes-Gallareta, Claudia Arce-Salinas, Oscar Arrieta, Luis L Rojas-Puentes, Marcelino Gonzalez-Pinedo, Alejando Crismatt, Alette Ortega-Gomez, Carlos Gamboa-Vignolle, Rodrigo Nuñez-Gomez, Yusmiren Dorantes-Gallareta, Claudia Arce-Salinas, Oscar Arrieta

Abstract

Background and purpose: Chloroquine (CLQ), an antimalarial drug, has a lysosomotropic effect associated with increased radiationsensibility, which is mediated by the leakage of hydrolytic enzymes, increased apoptosis, autophagy and increased oxidative stress in vitro. In this phase II study, we evaluated the efficacy and safety of radiosensibilization using CLQ concomitant with 30 Gray (Gy) of whole-brain irradiation (WBI) to treat patients with brain metastases (BM) from solid tumors.

Methods: Seventy-three eligible patients were randomized. Thirty-nine patients received WBI (30 Gy in 10 fractions over 2 weeks) concomitant with 150 mg of CLQ for 4 weeks (the CLQ arm). Thirty-four patients received the same schedule of WBI concomitant with a placebo for 4 weeks (the control arm). All the patients were evaluated for quality of life (QoL) using the EORTC Quality of Life (QoL) Questionnaire (EORTC QLQ-C30) (Mexican version) before beginning radiotherapy and one month later.

Results: The overall response rate (ORR) was 54% for the CLQ arm and 55% for the control arm (p=0.92). The progression-free survival of brain metastases (BMPFS) rates at one year were 83.9% (95% CI 69.4-98.4) for the CLQ arm and 55.1% (95% CI 33.6-77.6) for the control arm. Treatment with CLQ was independently associated with increased BMPFS (RR 0.31,95% CI [0.1-0.9], p=0.046).The only factor that was independently associated with increased overall survival (OS) was the presence of< 4 brain metastases (RR 1.9, 95% CI [1.12-3.3], p=0.017). WBI was associated with improvements in cognitive and emotional function but also with worsened nausea in both patients groups. No differences in QoL or toxicity were found between the study arms.

Conclusion: Treatment with CLQ plus WBI improved the control of BM (compared with the control arm) with no increase in toxicity; however, CLQ did not improve the RR or OS. A phase III clinical trial is warranted to confirm these findings.

Figures

Figure 1
Figure 1
Consort.
Figure 2
Figure 2
Efficacy of Chloroquine. A. Progression Free Survival of Brain Metastases. B. Overall Survival. C. Event Free Survival. D. Mortality related to brain disease progression.

References

    1. Davey P. Brain metastases: treatment options to improve outcomes. CNS Drugs. 2002;16(5):325–338. doi: 10.2165/00023210-200216050-00005.
    1. Sheehan JP, Sun M-H, Kondziolka D, Flickinger J, Lunsford LD. Radiosurgery for non-small cell lung carcinoma metastatic to the brain: long-term outcomes and prognostic factors influencing patient survival time and local tumor control. J Neurosurg. 2002;97(6):1276–1281. doi: 10.3171/jns.2002.97.6.1276.
    1. O’Neill BP, Iturria NJ, Link MJ, Pollock BE, Ballman KV, O’Fallon JR. A comparison of surgical resection and stereotactic radiosurgery in the treatment of solitary brain metastases. Int J Radiat Oncol Biol Phys. 2003;55(5):1169–1176. doi: 10.1016/S0360-3016(02)04379-1.
    1. Nathoo N, Chahlavi A, Barrett GH, Toms SA. Pathobiology of brain metastases. J ClinPathol. 2005;58:237–242.
    1. Jun X, Gang P, Jing-Song Y, Qian D, Jing C. Predictive factors of brain metastasis in patients with breast cancer. Med Oncol. 2013;30:337.
    1. Klos KJ, O’Neill BP. Brain metastases. Neurologist. 2004;10:31–46. doi: 10.1097/01.nrl.0000106922.83090.71.
    1. Langer CJ, Mehta MP. Current management of brain metastases, with a focus on systemic options. J ClinOncol. 2005;23:6207–6219. doi: 10.1200/JCO.2005.03.145.
    1. Patchell RA, Tibbs PA, Walsh JW, Dempsey RJ, Maruyama Y, Kryscio RJ, Markesbery WR, Macdonald JS, Young B. A randomized trial of surgery in the treatment of single metastasis to the brain. N Engl J Med. 1990;322(8):494–500. doi: 10.1056/NEJM199002223220802.
    1. Arrieta O, Villareal-Garza C, Zamora J, Blake-Cerda M, De La Mata M, Diego Zavala D, Muñiz-Hernández S, De La Garza J. Long-term survival in patients with non-small cell lung cancer and synchronous brain metastasis treated with whole. Brain radiotherapy and thoracic chemoradiation. Radiat Oncol. 2011;6:166. doi: 10.1186/1748-717X-6-166.
    1. Villareal-Garza C, De La Mata D, Zavala D, Macedo-Perez E, Arrieta O. Aggressive tretament of primary tumor in patients with non-small-cell lung cancer and exclusively brain. Metastasis. 2013;14(1):6–13.
    1. Mehta M, Paleologos N, Mikkelsen M, D.Robinson P, Ammirati M, Andrews D. The role of chemotherapy in the management of newly diagnosed brain metastasis: a systemic review and evidence-based clinical practice guideline. J Neurooncol. 2010;96:71–83. doi: 10.1007/s11060-009-0062-7.
    1. Gamboa-Vignolle C, Ferrari-Carballo T, Arrieta O, Mohar A. Whole-brain irradiation with concomitant daily fixed-dose Temozolomide for brain metastasis treatment: a randomized phase II trial. Radiother Oncol. 2012;2:187–191.
    1. Verger E, Gil M, Yaya R, Viñolas N, Villa S, Pujol T, Quintó L, Graus F. Temozolomide and concomitant whole brain radiotherapy in patients with brain metástasis: a phase II randomized trial. Int J RadiatOncolBiol Phys. 2005;61(1):185–191.
    1. Chua D, Krzakowski M, Chouaid C, Pallotta MG, Martinez JI, Gottfried M, Curran W, Throuvalas N. Whole brain radiation therapy plus concomitant temozolamide for the treatment of brain metastasis from non-small-cell lung cancer: a randomized, open-label phase II study. Clin Lung Cancer. 2010;11(3):176–181. doi: 10.3816/CLC.2010.n.022.
    1. Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloro- quine on viral infections: an old drug against today’s diseases? Lancet Infect Dis. 2003;3:722–727. doi: 10.1016/S1473-3099(03)00806-5.
    1. Giampietri A, Fioretti MC, Goldin A, Bonmassar E. Drug-mediated antigenic changes in murine leukemia cells: antagonistic effects of quinacrine, an antimutagenic compound. J Natl Cancer Inst. 1980;64:297–301.
    1. Kim EL, Wustenberg R, Rubsam A, Schmitz-Salue C, Warnecke G, Eva-Maria B, Pettkus N, Speidel D, Rohde V, Schultz-Schaeffer W, Deppert W, Giese A. Chloroquine activates the p53 pathway and induces apoptosis in human glioma cells. Neuro Oncol. 2010;12(4):389–400. doi: 10.1093/neuonc/nop046.
    1. Harhaji-Trajkovic L, Arsikin K, Kravic-Streovovic T, Petricevic S, Tovilovic G, Pantovic A. Chloroquine-mediated lysosomal dysfunction enhances the anticancer effect of nutrient deprivation. Pharm Res. 2012;29(8):2249–2263. doi: 10.1007/s11095-012-0753-1.
    1. Toler SM, Noe D, Sharma A. Selective enhancement of cellular oxidative stress by chloroquine:implications for the treatment of glioblastoma multiforme. Neurosurg Focus. 2006;21(6):E10.
    1. Sotelo J, Briceno E, López-González A. Adding chloroquine to conventional treatment for glioblastoma multiforme. Ann Intern Med. 2006;144(5):337–343. doi: 10.7326/0003-4819-144-5-200603070-00008.
    1. Briceño E, Calderon A, Sotelo J. Institutional experience with chloroquine as an adjuvant to the therapy for glioblastoma multiforme. Surg Neurol. 2007;67(4):388–391. doi: 10.1016/j.surneu.2006.08.080.
    1. Therasse P, Arbuck SG, Eisenhauer EA. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst. 2000;92:205–216. doi: 10.1093/jnci/92.3.205.
    1. Arrieta O, Núñez-Valencia C, Reynoso-Erazo L, Alvarado S, Flores-Estrada D, Angulo LP, Oñate-Ocaña L. Health-related quality of life in patients with lung cancer: validation of the Mexican-Spanish version and association with prognosis of the EORTC QLQ-LC13 questionnaire. Lung Cancer. in press.
    1. Briceño E, Reyes S, Sotelo J. Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine. [ ]
    1. Reyes S, Herrera LA, Ostrosky P, Sotelo J. Quinacrine enhances carmustine therapy of experimental rat glioma. Neurosurgery. 2001;49:969–973.
    1. Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a target for anticancer therapy. Nat Rev Clin Oncol. 2011;8:528–539. doi: 10.1038/nrclinonc.2011.71.
    1. Maclean KH, Dorsey FC, Cleveland JL, Kastan MB. Targeting lysosomal degradation induces p53-dependent cell death and prevents cancer in mouse models of lymphomagenesis. J Clin Invest. 2008;118:79–88. doi: 10.1172/JCI33700.
    1. Amaravadi RK. Autophagy inhibition enhances therapy-induced apoptosis in a Mycinduced model of lymphoma. J Clin Invest. 2007;117:326–336. doi: 10.1172/JCI28833.
    1. Carew JS. Autophagy inhibition enhances vorinostat-induced apoptosis via ubiquitinated protein accumulation. J Cell Mol Med. 2010;14:2448–2459. doi: 10.1111/j.1582-4934.2009.00832.x.
    1. Bellodi C. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest. 2009;119:1109–1123. doi: 10.1172/JCI35660.
    1. Carew JS. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood. 2007;110:313–322. doi: 10.1182/blood-2006-10-050260.
    1. Gupta A. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST) Proc Natl Acad Sci. 2010;107:14333–14338. doi: 10.1073/pnas.1000248107.
    1. Guerrieri M, Wong K, Ryan G, Millward M, Quong G, Ball DL. A randomised phase III study of palliative radiation with concomitant carboplatin for brain metastases from non-small cell carcinoma of the lung. Lung Cancer. 2004;46:107–111. doi: 10.1016/j.lungcan.2004.02.019.
    1. Arruda-Viani G, Borges-Manta G, Carrara Fonseca E, Issa De fendi L, Luis Alfonso S, Stefano EJ. Whole brain radiotherapy with radiosensitizer for brain metastases. J Exp Clin Cancer Res. 2009;28:1–11. doi: 10.1186/1756-9966-28-1.

Source: PubMed

3
Suscribir