603558

research-article2015

TAU0010.1177/1756287215603558Therapeutic Advances in UrologyA. R. Mehta and A. J. Armstrong

Therapeutic Advances in Urology

Review

Tasquinimod in the treatment of castrate-resistant prostate cancer – current status and future prospects

Ther Adv Urol 2016, Vol. 8(1) 9­–18 DOI: 10.1177/ 1756287215603558 © The Author(s), 2015. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav

Amit R. Mehta and Andrew J. Armstrong

Abstract:  Treatment options have significantly expanded in recent years for men with metastatic castration-resistant prostate cancer (mCRPC), with the routine use of immunotherapy (sipuleucel-T) and novel hormonal agents such as enzalutamide and abiraterone acetate prior to taxane-based chemotherapy or radium-223 radiotherapy. A number of immune checkpoints limit the immune response of the host to metastatic tumor progression in prostate cancer, one of which is an immunosuppressive and pro-angiogenic cell called the myeloid-derived suppressor cell (MDSC). Tasquinimod is a small molecular oral inhibitor of S100A9, a key cell surface regulator of MDSC function, and has shown antiangiogenic, antitumor and immune-modulatory properties in preclinical models of prostate cancer and other solid tumors. A large randomized phase II trial of tasquinimod in men with chemotherapy-naïve mCRPC demonstrated a significant prolongation in radiographic and symptomatic progression-free survival compared with placebo, which was also associated with improvements in overall survival. Tasquinimod was studied in a global phase III randomized trial in men with bone mCRPC and, while it significantly improved radiographic progression-free survival, this did not result in an overall survival benefit. However, tasquinimod is under evaluation as well as a combination therapy with other systemic agents in prostate cancer and as a single agent in other solid tumors. This review encompasses the preclinical and clinical development of tasquinimod as a therapy for men with prostate cancer.

Keywords:  angiogenesis, castration resistant, immunomodulatory, metastasis tasquinimod, myeloid derived suppressor cell, prostate cancer

Background Prostate carcinoma remains the most common noncutaneous malignancy in men in the United States, with 220,000 cases diagnosed in 2014 [Siegel et  al. 2014]. The estimated incidence of death from this disease is estimated at 27,540 patients annually, a number that continues to fall by 3–4% each year [Siegel et al. 2014]. Targeting the androgen receptor axis has been classically known as an effective strategy in recurrent and metastatic prostate adenocarcinoma, and novel anti-neoplastic drugs have been developed and approved to target the androgen receptor as well, including enzalutamide and abiraterone [Beer et al. 2014; de Bono et al. 2011]. However, patients eventually experience progression of disease in

spite of these newer androgen receptor axis targeting therapies and this has spurred the discovery of other mechanisms of action that have antineoplastic activity. These include immunologic therapies such as sipuleucel-T, ipilimumab and Prostvac, and antiangiogenic molecules such as cabozantinib [Thoreson et  al. 2014]. But despite novel hormonal agents, taxane chemotherapy and radium-223 therapy, men with metastatic castration-resistant prostate cancer (mCRPC) face a short survival of 1–3 years depending on context, illustrating the need for novel approaches to systemic therapy. One compound under active phase III investigation in prostate cancer, with a novel mechanism of action, is tasquinimod [Isaacs et al. 2006].

Correspondence to: Andrew J. Armstrong, MD, ScM, FACP Associate Professor of Medicine and Surgery, Associate Director for Clinical Research in Genitourinary Oncology, Duke Cancer Institute, Divisions of Medical Oncology and Urology, Duke University, DUMC Box 103861, Durham, NC 27710, USA andrew.armstrong@ duke.edu Amit R. Mehta, MD Duke Cancer Institute Genitourinary Program, Cary, NC, USA

http://tau.sagepub.com 9

Therapeutic Advances in Urology 8(1) Tasquinimod development Tasquinimod (Figure 1) was originally identified as a potential anticancer compound during a drug screening program for anti-inflammatory molecules and was observed to promote rather than inhibit inflammation [Isaacs, 2010]. This finding catalyzed the study of tasquinimod in humans, which was demonstrated to retain some proinflammatory activity but with reduced autoimmunity, but also retained potent antitumor activity in prostate cancer [Isaacs, 2010]. The primary antitumor mechanism of action is felt to be via engagement of the S100A9 protein, which is an immunomodulatory protein found in the tumor microenvironment and on myeloidderived suppressor cells [Bjork et  al. 2009; Hermani et al. 2005]. The S100A9 protein has been found to be a ligand for receptor for advanced glycation end products (RAGE) and to have a highly specific interaction with Toll-like receptor 4 (TLR-4) [Bjork et  al. 2009]. In general, the referenced myeloid-derived suppressor cells (MDSC) have their growth and phenotype induced by chronic inflammation, which is accomplished by signaling through TLR-4 [Bunt et al. 2009]. Preclinical work demonstrated that interleukin-1β inflammation led to MDSC producing increased interleukin-10, thereby promoting a type 2 immune response, which fosters tumor progression [Becker, 2006]. In prostate cancer specifically, the calcium-modulated proteins S100A8 and S100A9 along with RAGE are upregulated in both prostatic intraepithelial neoplasia and adenocarcinoma, whereas benign prostatic tissue showed minimal to no expression [Hermani et al. 2005]. In malignant prostate tissue, when S100A9 interacts and subsequently modulates RAGE, there is activation of multiple cellular pathways including mitogenactivated protein kinases (MAP kinase) and nuclear factor κB. This consequently directly affects cell survival, invasion/metastasis and inflammatory response [Taguchi et  al. 2000]. Furthermore, blockade of RAGE along with its colocalizing polypeptide amphoterin in mice with implanted tumors led to decreased cancer growth and metastases [Taguchi et  al. 2000]. Based on these series of findings, it has also been hypothesized that MDSC are a prospective inflammatory checkpoint to target and employment of tasquinimod may suppress MDSC, which subsequently may kindle an antitumor response [Bunt et al. 2009]

Figure 1.  Chemical structure of tasquinimod: 4-hydroxy-5-methoxy-N,1-dimethyl-2-oxo-N-[(4trifluoromethyl) phenyl]-1,2-dihydroquinoline-3carboxamide).

Preclinical studies have also shown that tasquinimod modulates the anti-angiogenic molecule thrombospondin-1 in prostate tumor LNCaP cells [Olsson et  al. 2010]. Following exposure to tasquinimod, thrombospondin-1 mRNA was demonstrated to be upregulated. In the same series of experiments, human prostate tumors implanted in mice were also administered tasquinimod and a multitude of effects were seen: (1) tumor growth inhibition; (2) upregulation of thrombospondin-1; (3) downregulation of hypoxia-inducible factor 1α (HIF-1α); and (4) downregulation of androgen receptor. Furthermore, evidence of antiangiogenic activity was seen, as tumor vascular endothelial growth factor (VEGF) levels were either stable or decreased upon treatment with tasquinimod [Olsson et  al. 2010]. In addition, thrombospondin-1 also inhibits neovascularization via blockade of VEGF receptors on endothelial cells [Doll et al. 2001]. In addition, tasquinimod has demonstrated antiangiogenic properties in a number of vascular assays, including VEGF-independent mechanisms. While the exact mechanisms remain unclear, there are data supporting its role in inhibiting the angiogenic switch mediated by tumor hypoxia, perhaps by regulating epigenetic effectors of angiogenesis such as histone deacetylase (HDAC) 3/4 [Isaacs et al. 2012]. In addition, MDSCs are known to express S100A9 and are pro-angiogenic. This immunosuppressive cellular checkpoint is increased in many solid tumor settings, including prostate cancer, and thus tasquinimod’s anti-angiogenic effects may in part be mediated through an indirect immunomodulatory function and inhibition of MDSC activity. Put together, tasquinimod has a unique antineoplastic mechanism, via both direct antiproliferation and antitumor properties perhaps triggered

10 http://tau.sagepub.com

AR Mehta and AJ Armstrong through engagement of the S100A9 protein, as well as immune modulatory and indirect antiangiogenic effects, in prostate cancer compared with other compounds currently available in the clinic. Preclinical data The development of tasquinimod was launched via studies in rat prostate cancer models, spearheaded by the Isaacs laboratory at Johns Hopkins University in conjunction with Active Biotech in Sweden. The predecessor molecule to tasquinimod, named linomide (4-hydroxy-N,1dimethyl2-oxo-N-phenyl-1,2-dihydroquinoline-3-carboxamide), had shown antineoplastic effects via antiangiogenic activity in prostate cancer xenografts, but further study with linomide was terminated due to autoimmune and proinflammatory and cardiovascular toxicity [Ichikawa et al. 1992; Noseworthy et al. 2000]. On account of these promising anticancer properties, however, 11 derivative quinolone-3-carboxamide compounds were tested to see if one could be found that retained antiangiogenic ability but without the deleterious proinflammatory stimulation [Isaacs et  al. 2006]. Chemically, linomide was modified by way of substitutions in the aromatic rings. Then, each of these molecules was tested in vivo to measure the inhibition of prostate cancer growth in the Dunning R-3327 AT-1 rat model. In addition, each molecule’s potency in inhibiting angiogenesis was also was analyzed through in vivo assays in mice. The results from these two series of experiments allowed for a ranking of the 11compounds, and the one that emerged as the lead compound was ABR-215050 [4-hydroxy-5-methoxy-N,1-dimethyl2-oxo-N-[(4-trifluoromethyl) phenyl]-1,2-dihydroquinoline-3-caboxamide]; this was later named tasquinimod. The next critical step in the preclinical phase was to examine the inflammatory properties of ABR215050, because the proinflammatory aspects of linomide mediated the toxicities leading to its discontinuation. ABR-215050 (tasquinimod) was assessed in a Beagle dog model validated for measuring inflammatory response and a lack of proinflammatory effect was observed [Snyder et al. 1995]. Specifically, after intravenous injections of 1 mg ABR215050/kg/day for 5 days, white blood cell count (increase of 800/ml) and erythrocyte sedimentation rate (no increase) results supported this absence of induced

inflammation in this model [Isaacs et  al. 2006]. This further provided support for continuing development. ABR-215050 then underwent robust characterization of its dose–response ability to abrogate prostate cancer growth in a total of four human and rodent prostate cancer models, as well as additional assays to measure antiangiogenic proficiency. To measure the potency of inhibiting cancer growth, the total daily dose that inhibits tumor by 50% (ED50 concentration) was calculated. Based on the ED50 values for both the human and rodent prostate cancer models, ABR-215050 is approximately 30–60 times as efficacious as linomide [Isaacs et al. 2006]. In the four angiogenesis assays studied, results suggested at least a 50% inhibition of angiogenesis in the presence of ABR-215050, independent of effects on the immune components of the tumor microenvironment. For example, in the in vivo CAM assay in chicken eggs, 10 µM of ABR215050 was co-administered with 100 ng of VEGF. At 48 h, the angiogenic response decreased by more than 50% [Isaacs et al. 2006]. Importantly, among the human and rodent prostate cancer models, it was observed that degree of antineoplastic effect was proportional to the antiangiogenic activity. For example, in the models examined in the Isaacs lab at Johns Hopkins, a tumor blood vessel density diminution of 44–65% corresponded to a 41–78% reduction in tumor weight. To further investigate the antiangiogenic mechanism, tumor tissue oxygenation was also measured. CWR-22Rv1 tumor bearing mice were given either 10 mg ABR-215050/kg/day versus nothing for 1 month. Subsequently, utilizing a fiberoptic probe placed in the center of tumors, the partial pressure of oxygen (pO2) was recorded. It was found that in treated tumors, the mean pO2 was 5.6 mmHg compared with 15.1 mmHg in untreated tumors. Statistical analysis calculated a p value of

Tasquinimod in the treatment of castrate-resistant prostate cancer - current status and future prospects.

Treatment options have significantly expanded in recent years for men with metastatic castration-resistant prostate cancer (mCRPC), with the routine u...
458KB Sizes 1 Downloads 12 Views