scispace - formally typeset
Search or ask a question
Author

Kitty Wan

Bio: Kitty Wan is an academic researcher from Novartis. The author has contributed to research in topics: Medicine & Pharmacodynamics. The author has an hindex of 3, co-authored 3 publications receiving 54 citations.

Papers
More filters
Journal ArticleDOI
TL;DR: In ER+ BC xenograft models, LEE011 has demonstrated enhanced antitumor activity when combined with BYL719 and letrozole versus each agent alone, suggesting this combination may optimize treatment in ER+BC.
Abstract: 533 Background: Postmenopausal women with advanced ER+ BC are usually treated with aromatase inhibitors (AIs) as first-line therapy but resistance eventually develops. Dysregulation of the PI3K pat...

33 citations

Proceedings ArticleDOI
TL;DR: The dose escalation results from a Ph I-Ib/II study of MBG453 ± spartalizumab in metastatic solid tumors show synergistic anti-tumor activity of TIM-3 and PD-1 co-blockade generally increased in a dose-proportional manner.
Abstract: Background: MBG453 and spartalizumab, humanized IgG4 mAbs, block binding of TIM-3 to PtdSer and PD-1 to PD-L1/2, respectively. Preclinical studies show synergistic anti-tumor activity of TIM-3 and PD-1 co-blockade. Here we report the dose escalation results from a Ph I-Ib/II study of MBG453 ± spartalizumab in metastatic solid tumors (NCT02608268). Methods: Metastatic cancer pts received intravenous MBG453 alone at 80-1200 mg Q2W or Q4W, or combination therapy (Q2W/Q4W) with MBG453 (20-800/80-1200 mg) + spartalizumab (80-240 mg/80-400 mg). The recommended Ph II dose (RP2D) was determined using an adaptive Bayesian logistic regression model guided by escalation with overdose control together with Ph I-Ib endpoints (dose-limiting toxicity [DLT] at 8 weeks). Results: As of July 26, 2018, 87 pts received MBG453 alone (14% were anti-PD-1/PD-L1 pre-treated; common tumors [≥9%]: pancreatic cancer [11%], sarcoma [10%], and colorectal cancer [CRC; 9%]) and 86 pts received MBG453 + spartalizumab (27% were anti-PD-1/PD-L1 pre-treated; common tumors [≥6%]: melanoma [8%], NSCLC [7%], CRC [7%], and ovarian cancer [6%]). One DLT (myasthenia gravis grade [G] 4) was reported in a pt with thymoma treated with MBG453 (240 mg Q4W) + spartalizumab (80 mg Q4W). Treatment-related adverse events (AEs) were reported in 40% and 59% of pts treated with MBG453 and MBG453 + spartalizumab, and G3/4 AEs in 0% and 11% of pts; the most common AE (≥10%) was fatigue in 10% and 15% of pts, respectively. MBG453 exposure generally increased in a dose-proportional manner. Maximum tolerated doses were not identified with the tested dose/schedule. MBG453 800 mg Q4W (n=9 pts treated) and MBG453 800 mg + spartalizumab 400 mg Q4W (n=6 pts treated) were declared as RP2Ds. Stable disease (SD) was seen in 25/87 (29%) pts treated with MBG453 alone (common tumors [≥2 pts]: sarcoma [n=5], and breast cancer, CRC, ovarian cancer, cholangiocarcinoma, and NSCLC [n=2 pts each]); 4 of 25 pts with SD were anti-PD-1/PD-L1 pre-treated. Of 86 MBG453 + spartalizumab pts, partial responses were seen in 4 pts (5%); 1 anti-PD-1/PD-L1 pre-treated pt (out of 6 with NSCLC; DOR: 392 d), 3 anti-PD-1/PD-L1 naive pts (CRC [n=2 out of 6, DOR: 223 d, 109 d]; SCLC [n=1 out of 3; DOR: 112 d]). SD was seen in 34/86 (40%) MBG453 + spartalizumab pts (common tumors [≥3 pts]: melanoma [n=5; 2 cutaneous, 2 uveal, and 1 non-cutaneous], ovarian cancer [n=3], and urothelial carcinoma [n=3]); 10 of 34 pts with SD were anti-PD-1/PD-L1 pre-treated. RNAseq analysis of screening and on-treatment biopsies revealed a pharmacodynamic trend of increased IFN-γ-associated gene signatures following MBG453 + spartalizumab treatment. Conclusions: MBG453 + spartalizumab was well tolerated with preliminary signs of anti-tumor activity. MBG453 800 mg Q4W and MBG453 800 mg + spartalizumab 400 mg Q4W were selected as the RP2Ds; dose expansion is ongoing in pts with melanoma or NSCLC resistant to anti-PD-1/PD-L1. Citation Format: Giuseppe Curigliano, Hans Gelderblom, Nicolas Mach, Toshihiko Doi, Wai Meng David Tai, Patrick Forde, John Sarantopoulos, Philippe L. Bedard, Chia-Chi Lin, Stephen Hodi, Sofie Wilgenhof, Armando Santoro, Catherine Sabatos-Peyton, Tyler Longmire, Kitty Wan, Panagiotis Nikolopoulos, Luigi Manenti, Aung Naing. Phase (Ph) I/II study of MBG453± spartalizumab (PDR001) in patients (pts) with advanced malignancies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT183.

25 citations

Journal ArticleDOI
TL;DR: In this paper, the adaptive scaled independence sampler (ASIS) algorithm is applied to logistic regression and accelerated failure time models, and compared with data augmentation, Laplace approximation and correlated pseudo-marginal method.
Abstract: Bayesian variable selection is an important method for discovering variables which are most useful for explaining the variation in a response. The widespread use of this method has been restricted by the challenging computational problem of sampling from the corresponding posterior distribution. Recently, the use of adaptive Monte Carlo methods has been shown to lead to performance improvement over traditionally used algorithms in linear regression models. This paper looks at applying one of these algorithms (the adaptively scaled independence sampler) to logistic regression and accelerated failure time models. We investigate the use of this algorithm with data augmentation, Laplace approximation and the correlated pseudo-marginal method. The performance of the algorithms is compared on several genomic data sets.

7 citations

Journal ArticleDOI
TL;DR: In this article , the safety, tolerability, and preliminary antitumor activity of naporafenib (LXH254), a BRAF/CRAF protein kinases inhibitor, were explored in combination with trametinib in patients with NRAS-mutant melanoma.
Abstract: PURPOSE No approved targeted therapy for the treatment of patients with neuroblastoma RAS viral (v-ras) oncogene homolog (NRAS)–mutant melanoma is currently available. PATIENTS AND METHODS In this phase Ib escalation/expansion study (ClinicalTrials.gov identifier: NCT02974725), the safety, tolerability, and preliminary antitumor activity of naporafenib (LXH254), a BRAF/CRAF protein kinases inhibitor, were explored in combination with trametinib in patients with advanced/metastatic KRAS- or BRAF-mutant non–small-cell lung cancer (escalation arm) or NRAS-mutant melanoma (escalation and expansion arms). RESULTS Thirty-six and 30 patients were enrolled in escalation and expansion, respectively. During escalation, six patients reported grade ≥3 dose-limiting toxicities, including dermatitis acneiform (n = 2), maculopapular rash (n = 2), increased lipase (n = 1), and Stevens-Johnson syndrome (n = 1). The recommended doses for expansion were naporafenib 200 mg twice a day plus trametinib 1 mg once daily and naporafenib 400 mg twice a day plus trametinib 0.5 mg once daily. During expansion, all 30 patients experienced a treatment-related adverse event, the most common being rash (80%, n = 24), blood creatine phosphokinase increased, diarrhea, and nausea (30%, n = 9 each). In expansion, the objective response rate, median duration of response, and median progression-free survival were 46.7% (95% CI, 21.3 to 73.4; 7 of 15 patients), 3.75 (95% CI, 1.97 to not estimable [NE]) months, and 5.52 months, respectively, in patients treated with naporafenib 200 mg twice a day plus trametinib 1 mg once daily, and 13.3% (95% CI, 1.7 to 40.5; 2 of 15 patients), 3.75 (95% CI, 2.04 to NE) months, and 4.21 months, respectively, in patients treated with naporafenib 400 mg twice a day plus trametinib 0.5 mg once daily. CONCLUSION Naporafenib plus trametinib showed promising preliminary antitumor activity in patients with NRAS-mutant melanoma. Prophylactic strategies aimed to lower the incidence of skin-related events are under investigation.

2 citations

Journal ArticleDOI
TL;DR: Evaluated safety and tolerability, and determine the recommended dose for expansion (RDE), and characterize pharmacokinetics and pharmacodynamics of LXH254 + TMT, and evaluate antitumor activity.
Abstract: Background: MAPK pathway dysregulation is a characteristic molecular abnormality of melanoma. NRAS mutations in melanoma lead to constitutive MAPK activation and subsequent MEK and ERK activation. Inhibiting the pathway with a combination of BRAF/CRAF and MEK inhibitors (LXH254 and TMT, respectively) has demonstrated synergistic activity in preclinical models of NRAS-mutant melanoma. Methods: We report data from an open-label, Phase Ib study (NCT02974725), that tested LXH254 + TMT combination in a dose-escalation (ESC) part in pts with KRAS- or BRAF-mutant NSCLC or NRAS-mutant melanoma and in a dose-expansion (EXP) part in NRAS-mutant melanoma pts. In ESC, five dose levels were explored: LXH254 200 mg twice daily (BID) + TMT (1 mg or 0.5 mg) daily (QD); LXH254 400 mg BID + TMT (1 mg or 0.5 mg) QD; LXH254 400 mg BID + TMT 1 mg (QD, 2 wk on/2 wk off). Primary objectives were to evaluate safety and tolerability, and determine the recommended dose for expansion (RDE). Secondary objectives were to characterize pharmacokinetics and pharmacodynamics of LXH254 + TMT, and evaluate antitumor activity. Results: As of Dec 9, 2021, 36 and 30 pts were enrolled in ESC and EXP, respectively; median age was 63.5 and 69 y, respectively. Pts in ESC and EXP received a median of 3 (range: 1-6) and 2 (range: 1-7) prior treatments, respectively, including IO (ESC: 88.9% pts; EXP: 96.7% pts). All pts from ESC and 24 pts from EXP discontinued, mainly due to progressive disease (61.1% and 60%, respectively); 6 pts were ongoing in EXP at data cut-off. In ESC, Grade ≥3 (G≥3) DLTs were reported in 6 pts: dermatitis acneiform (n=2), maculopapular rash (n=2), increased lipase (n=1), and Stevens-Johnson syndrome (n=1). The RDEs were LXH254 200 mg BID + TMT 1 mg QD and LXH254 400 mg BID + TMT 0.5 mg QD. Treatment-related adverse events (TRAEs) were reported in 34 pts (94.4%) in ESC and all pts in EXP. G≥3 TRAEs in ESC and EXP were experienced by 19 (52.8%) and 24 (80%) pts, respectively; the most common being rash or dermatitis acneiform (13.9% each) in ESC and rash (33.3%) in EXP. One fatal TRAE (hypovolemic shock) was reported in the LXH254 400 mg BID + TMT 0.5 mg QD group. The ORR in EXP was 46.7% (7 pts; 95% CI: 21.3-73.4) for LXH254 200 mg BID + TMT 1 mg QD and 13.3% (2 pts; 95% CI: 1.7-40.5) for LXH254 400 mg BID + TMT 0.5 mg QD; overall in EXP, 9 pts (30%) achieved PR and 13 pts (43.3%) reported SD. In EXP the median DOR was 3.8 months (95% CI: 2.6-7.4). Exposures of LXH254 (200 mg or 400 mg BID) + TMT (0.5 mg or 1 mg QD) vs LXH254 single agent were comparable, indicating no apparent LXH254-TMT interactions. Limited paired tumor biopsy data (n=8) showed substantial MAPK pathway inhibition (DUSP6). Conclusions: LXH254 in combination with TMT has shown preliminary antitumor activity in the pretreated, NRAS-mutant melanoma population. A Phase II trial of LXH254 combinations in pts with pretreated BRAF- or NRAS-mutant melanoma is currently ongoing (NCT04417621). Citation Format: Filippo de Braud, Christophe Dooms, Rebecca S. Heist, Celeste Lebbe, Martin Wermke, David Planchard, Dirk Schadendorf, Piotr Rutkowski, Jürgen Wolf, Paolo A. Ascierto, Ignacio Gil-Bazo, Shumei Kato, Maria Wolodarski, Meredith McKean, Eva Muñoz Couselo, Martin Sebastian, Armando Santoro, Vesselina Cooke, Luca Manganelli, Kitty Wan, Anil Gaur, Tanay S. Samant, Jaeyeon Kim, Giordano Caponigro, Xuân-Mai Couillebault, Michele Moschetta, Adil Daud. Phase Ib study of LXH254 + trametinib (TMT) in patients (pts) with NRAS-mutant melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT197.

Cited by
More filters
Journal ArticleDOI
TL;DR: How CDK inhibitors with high selectivity (particularly for both CDK4 and CDK6), in combination with patient stratification, have resulted in more substantial clinical activity is discussed.
Abstract: Cancer represents a pathological manifestation of uncontrolled cell division; therefore, it has long been anticipated that our understanding of the basic principles of cell cycle control would result in effective cancer therapies. In particular, cyclin-dependent kinases (CDKs) that promote transition through the cell cycle were expected to be key therapeutic targets because many tumorigenic events ultimately drive proliferation by impinging on CDK4 or CDK6 complexes in the G1 phase of the cell cycle. Moreover, perturbations in chromosomal stability and aspects of S phase and G2/M control mediated by CDK2 and CDK1 are pivotal tumorigenic events. Translating this knowledge into successful clinical development of CDK inhibitors has historically been challenging, and numerous CDK inhibitors have demonstrated disappointing results in clinical trials. Here, we review the biology of CDKs, the rationale for therapeutically targeting discrete kinase complexes and historical clinical results of CDK inhibitors. We also discuss how CDK inhibitors with high selectivity (particularly for both CDK4 and CDK6), in combination with patient stratification, have resulted in more substantial clinical activity.

1,276 citations

Journal ArticleDOI
TL;DR: Results of pivotal phase III trials investigating palbociclib in patients with advanced-stage oestrogen receptor (ER)-positive breast cancer have demonstrated a substantial improvement in progression-free survival, with a well-tolerated toxicity profile.
Abstract: Uncontrolled cellular proliferation, mediated by dysregulation of the cell-cycle machinery and activation of cyclin-dependent kinases (CDKs) to promote cell-cycle progression, lies at the heart of cancer as a pathological process. Clinical implementation of first-generation, nonselective CDK inhibitors, designed to inhibit this proliferation, was originally hampered by the high risk of toxicity and lack of efficacy noted with these agents. The emergence of a new generation of selective CDK4/6 inhibitors, including ribociclib, abemaciclib and palbociclib, has enabled tumour types in which CDK4/6 has a pivotal role in the G1-to-S-phase cell-cycle transition to be targeted with improved effectiveness, and fewer adverse effects. Results of pivotal phase III trials investigating palbociclib in patients with advanced-stage oestrogen receptor (ER)-positive breast cancer have demonstrated a substantial improvement in progression-free survival, with a well-tolerated toxicity profile. Mechanisms of acquired resistance to CDK4/6 inhibitors are beginning to emerge that, although unwelcome, might enable rational post-CDK4/6 inhibitor therapeutic strategies to be identified. Extending the use of CDK4/6 inhibitors beyond ER-positive breast cancer is challenging, and will likely require biomarkers that are predictive of a response, and the use of combination therapies in order to optimize CDK4/6 targeting.

760 citations

Journal ArticleDOI
TL;DR: Rapidly emerging data with selective inhibitors of CDK4/6 have validated these cell-cycle kinases as anticancer drug targets, corroborating longstanding preclinical predictions.
Abstract: Biochemical and genetic characterization of D-type cyclins, their cyclin D–dependent kinases (CDK4 and CDK6), and the polypeptide CDK4/6 inhibitor p16INK4 over two decades ago revealed how mammalian cells regulate entry into the DNA synthetic (S) phase of the cell-division cycle in a retinoblastoma protein–dependent manner. These investigations provided proof-of-principle that CDK4/6 inhibitors, particularly when combined with coinhibition of allied mitogen-dependent signal transduction pathways, might prove valuable in cancer therapy. FDA approval of the CDK4/6 inhibitor palbociclib used with the aromatase inhibitor letrozole for breast cancer treatment highlights long-sought success. The newest findings herald clinical trials targeting other cancers. Significance: Rapidly emerging data with selective inhibitors of CDK4/6 have validated these cell-cycle kinases as anticancer drug targets, corroborating longstanding preclinical predictions. This review addresses the discovery of these CDKs and their regulators, as well as translation of CDK4/6 biology to positive clinical outcomes and development of rational combinatorial therapies. Cancer Discov; 6(4); 353–67. ©2015 AACR .

682 citations

Journal ArticleDOI
TL;DR: The combination of letrozole and alpelisib was safe, with reversible toxicities, and clinical activity was observed independently of Pik3CA mutation status, although clinical benefit was seen in a higher proportion of patients with PIK3CA-mutated tumors.
Abstract: Purpose Alpelisib, a selective oral inhibitor of the class I PI3K catalytic subunit p110α, has shown synergistic antitumor activity with endocrine therapy against ER+/PIK3CA-mutated breast cancer cells. This phase Ib study evaluated alpelisib plus letrozole's safety, tolerability, and preliminary activity in patients with metastatic ER+ breast cancer refractory to endocrine therapy. Experimental design Twenty-six patients received letrozole and alpelisib daily. Outcomes were assessed by standard solid-tumor phase I methods. Tumor blocks were collected for DNA extraction and next-generation sequencing. Results Alpelisib's maximum-tolerated dose (MTD) in combination with letrozole was 300 mg/d. Common drug-related adverse events included hyperglycemia, nausea, fatigue, diarrhea, and rash with dose-limiting toxicity occurring at 350 mg/d of alpelisib. The clinical benefit rate (lack of progression ≥6 months) was 35% (44% in patients with PIK3CA-mutated and 20% in PIK3CA wild-type tumors; 95% CI, 17%-56%), including five objective responses. Of eight patients remaining on treatment ≥12 months, six had tumors with a PIK3CA mutation. Among evaluable tumors, those with FGFR1/2 amplification and KRAS and TP53 mutations did not derive clinical benefit. Overexpression of FGFR1 in ER+/PIK3CA mutant breast cancer cells attenuated the response to alpelisib in vitro CONCLUSIONS: The combination of letrozole and alpelisib was safe, with reversible toxicities. Clinical activity was observed independently of PIK3CA mutation status, although clinical benefit was seen in a higher proportion of patients with PIK3CA-mutated tumors. Phase II and III trials of alpelisib and endocrine therapy in patients with ER+ breast cancer are ongoing. Clin Cancer Res; 23(1); 26-34. ©2016 AACR.

259 citations

Journal ArticleDOI
TL;DR: The role of the different immune landscapes in CRC and their relationships with defined CRC genetic subtypes are discussed and in which ways CRC cells develop mechanisms to resist ICI are considered.
Abstract: Colorectal cancer (CRC) is highly heterogeneous at the genetic and molecular level, which has major repercussions on the efficacy of immunotherapy. A small subset of CRCs exhibit microsatellite instability (MSI), a molecular indicator of defective DNA mismatch repair (MMR), but the majority are microsatellite-stable (MSS). The high tumor mutational burden (TMB) and neoantigen load in MSI tumors favors the infiltration of immune effector cells, and antitumor immune responses within these tumors are strong relative to their MSS counterparts. MSI has emerged as a major predictive marker for the efficacy of immune checkpoint blockade over the last few years and nivolumab or pembrolizumab targeting PD-1 has been approved for patients with MSI refractory or metastatic CRC. However, some MSS tumors show DNA polymerase epsilon (POLE) mutations that also confer a very high TMB and may also be heavily infiltrated by immune cells making them amenable to respond to immune checkpoint inhibitors (ICI). In this review we discuss the role of the different immune landscapes in CRC and their relationships with defined CRC genetic subtypes. We discuss potential reasons why immune checkpoint blockade has met with limited success for the majority of CRC patients, despite the finding that immune cell infiltration of primary non-metastatic tumors is a strong predictive, and prognostic factor for relapse and survival. We then consider in which ways CRC cells develop mechanisms to resist ICI. Finally, we address the latest advances in CRC vaccination and how a personalized neoantigen vaccine strategy might overcome the resistance of MSI and MSS tumors in patients for whom immune checkpoint blockade is not a treatment option.

235 citations