Workflow
Arvinas LLC(ARVN) - 2025 Q3 - Quarterly Report

Clinical Development - The company is progressing multiple product candidates through clinical development, including ARV-102 for neurodegenerative disorders and ARV-393 for non-Hodgkin lymphoma [120]. - The ongoing Phase 1 clinical trial of ARV-102 in patients with Parkinson's disease has completed the single ascending dose cohort and is progressing to multiple ascending doses [124]. - The company plans to present initial data from the multiple dose cohort of the Phase 1 clinical trial of ARV-102 in patients with Parkinson's disease in 2026 [133]. - The Phase 1 clinical trial of ARV-102 included a randomized, double-blind, placebo-controlled design to assess safety and pharmacodynamics [125]. - The company has received Clinical Trial Application approval in the Netherlands for the Phase 1 clinical trial of ARV-102 in patients with Parkinson's disease [124]. - The Phase 1 clinical trial of ARV-102 was well tolerated, with no serious adverse events reported among 15 patients treated [136]. - The Phase 1 clinical trial of ARV-393 in patients with relapsed/refractory NHL is ongoing, with multiple responses observed in early cohorts [140]. - The company intends to share updated clinical data from the ongoing Phase 1 trial of ARV-393 at a medical congress in 2026 [140]. - The company plans to initiate a Phase 1 clinical trial of ARV-393 in combination with glofitamab in patients with DLBCL in 2026 [139]. - The company plans to initiate a first-in-human Phase 1 clinical trial for ARV-027 in healthy volunteers in 2026, pending regulatory feedback [166]. Product Efficacy - ARV-102 demonstrated over 90% reduction of LRRK2 in peripheral blood mononuclear cells at single doses of 60 mg or greater [131]. - In the Phase 1 clinical trial of ARV-102, treatment resulted in median LRRK2 protein reductions of 86% with a 50 mg dose and 97% with a 200 mg dose in patients with Parkinson's disease [136]. - ARV-393 showed strong synergistic antitumor activity in preclinical models of aggressive diffuse large B-cell lymphoma when combined with standard of care chemotherapy [135]. - ARV-393 in combination with R-CHOP chemotherapy achieved complete tumor regressions in all treated mice, indicating significant tumor growth inhibition (TGI) compared to other treatments [137]. - In preclinical studies, ARV-393 demonstrated robust tumor growth inhibition (≥95%) in two patient-derived xenograft (PDX) models of transformed follicular lymphoma (tFL) and showed enhanced antitumor activity in combination with small molecule inhibitors [141]. - ARV-806, a novel PROTAC KRAS G12D degrader, exhibited in vitro potency approximately 25 times greater than existing KRAS inhibitors and 40 times greater than the leading clinical-stage degrader [143]. - ARV-806 achieved over 90% degradation of KRAS G12D for seven days in a colorectal tumor xenograft model, indicating sustained pharmacodynamic activity [146]. - Vepdegestrant demonstrated near-complete ER degradation in tumor cells and superior anti-tumor activity compared to fulvestrant in preclinical studies [149]. Financial Performance - The company has raised approximately $1.7 billion in gross proceeds since inception through September 30, 2025, and received $913.1 million from collaboration partners and licensing arrangements [170]. - Revenue for Q3 2025 was $41.9 million, a decrease of $60.5 million from $102.4 million in Q3 2024, primarily due to reduced revenue from Novartis agreements [224]. - For the nine months ended September 30, 2025, revenue totaled $253.1 million, an increase of $48.9 million compared to $204.2 million in the same period of 2024, driven by a $157.9 million increase from the Vepdegestrant collaboration with Pfizer [225]. - The company recognized $20.0 million in revenue during the three months ended September 30, 2025, upon achieving a development milestone under the Novartis License Agreement [192]. - The company has not generated any revenue from product sales and does not expect to do so in the near future, relying instead on research collaborations and licensing arrangements [179]. Expenses and Cost Management - Research and development expenses for the three months ended September 30, 2025, totaled $64.7 million, a decrease of 25.5% compared to $86.9 million for the same period in 2024 [205]. - Total program-specific external expenses for the nine months ended September 30, 2025, were $89.2 million, down from $101.7 million in 2024, reflecting a decrease of 12.4% [205]. - The total unallocated internal expense for research and development was $24.4 million for the three months ended September 30, 2025, compared to $39.2 million in 2024, a decrease of 37.5% [205]. - General and administrative expenses for Q3 2025 totaled $21.0 million, a decrease of $54.8 million from $75.8 million in Q3 2024, mainly due to a loss on lease termination [230]. - The company expects to achieve annual operating cost savings of $100 million on a run-rate basis following workforce reductions [175]. - The company announced a workforce reduction of approximately 33% in April 2025 and an additional 15% in September 2025 to streamline operations [172][174]. Collaborations and Agreements - The collaboration agreement with Pfizer includes an upfront payment of $650 million and potential contingent payments of up to $1.4 billion based on regulatory and sales milestones [181]. - The Vepdegestrant (ARV-471) Collaboration Agreement allows for shared research and development expenses with Pfizer, which have been equal since July 22, 2021 [206]. - The Novartis Transaction resulted in a one-time upfront payment of $150.0 million and potential contingent payments of up to $1.01 billion based on specified milestones for luxdegalutamide (ARV-766) [192]. - The Bayer Collaboration Agreement was terminated effective August 12, 2024, after receiving a total of $29.5 million in upfront and additional payments [197][196]. - The company is eligible to receive up to $225.0 million in development milestone payments and up to $550.0 million in sales-based milestone payments under the Pfizer Research Collaboration Agreement [188]. Tax and Cash Management - As of December 31, 2024, the company had $111.0 million in federal net operating loss carryforwards, which can be carried forward indefinitely, subject to an 80% limit on taxable income [213]. - The company has provided a valuation allowance against deferred tax assets, indicating that it is more likely than not that the benefits will not be realized based on its earnings history [214]. - Cash, cash equivalents, and marketable securities amounted to $787.6 million as of September 30, 2025, compared to $1.0 billion as of December 31, 2024 [243]. - Net cash used in operating activities for the nine months ended September 30, 2025, increased by $68.2 million to $(243.4) million, primarily due to a $187.6 million decrease in deferred revenue [246]. - The company anticipates that its cash, cash equivalents, and marketable securities will fund its planned operating expenses into the second half of 2028 [252].