Common FDA Feedback in Type B Meeting Minutes for Solid Tumor IND Preparation

By Rhizome Team

Type B meetings with the FDA are critical milestones in the drug development process, providing sponsors with an opportunity to receive formal feedback on key development questions before filing an Investigational New Drug (IND) application. For therapies targeting solid tumors, these pre-IND meetings help identify potential issues and ensure alignment with FDA expectations.

Understanding common FDA feedback themes from Type B meeting minutes for solid tumor therapies can help sponsors better prepare their IND submissions, avoid delays, and design more robust development programs. The FDA's comments often focus on preclinical data requirements, clinical trial design, safety assessments, and manufacturing considerations.

Here we examine actual FDA feedback from Type B meeting minutes for solid tumor therapies preparing to file INDs, identifying recurring themes and specific issues raised by the agency. This analysis provides practical guidance for oncology drug developers navigating the pre-IND consultation process.

Want to read the citations or ask Rhizome a follow-up question? Schedule a call

what is common FDA feedback in type b meeting minutes for therapies targeting solid tumors when they are preparing to file their IND?

Answer

Executive synthesis

Across Type B meetings for solid tumor programs, FDA feedback commonly clusters into: (1) dose justification and quantitative clinical pharmacology (exposure–response, PK/PD modeling, DDI, QT/QTc) to support initial clinical dosing; (2) clinical trial design and endpoint expectations (e.g., BICR-assessed ORR/DOR, PFS acceptability and multiplicity control, durability of response for accelerated approval, early incorporation of PROs/COAs); (3) patient population clarity and representativeness (eligibility definitions, stratification factors, U.S. relevance); (4) safety monitoring and analysis plans (AESIs, narratives, pooled analyses); (5) standardized data and submission logistics to facilitate BIMO inspections; (6) CMC readiness (complete facility lists, stability packages, starting materials, control strategies, and, where applicable, device usability); (7) pediatric planning and, for biomarker-selected therapies, companion diagnostic alignment; and (8) targeted requests for additional studies or analyses (e.g., DDI, hepatic impairment, chronic tox, human factors) when evidence is insufficient. These themes recur across sponsors and tumor types and are typically expected to be addressed or well underway by IND filing or early in IND development 130132224529730048931391239242241234366482403996517572684231993884913623652232301989228019019238538127421910810424262654065215418051331017013494914174286369209202174533.

Dose selection and quantitative clinical pharmacology

FDA routinely asks sponsors to underpin proposed clinical doses with integrated quantitative analyses:

  • Exposure–response for efficacy and safety, PK/PD modeling, and simulation to justify dose and regimen, often leveraging longitudinal tumor growth models and baseline covariates 130132. FDA also expects complete bioanalytical method validation and final study reports with consistent subject IDs for clinical pharmacology data 224297300.
  • Cardiac safety and QT/QTc characterization, including concentration–QT modeling and submission of ECG waveforms to central warehouses where applicable 5256.
  • Drug–drug interactions and transporter considerations: examples include requests to use a strong CYP1A2 inhibitor (fluvoxamine) instead of ciprofloxacin for DDI assessment, and calculation of R-values to determine the need for clinical interaction studies with renal transporters (P-gp/BCRP/OCT1/MATE) 417414291.
  • Intrinsic/extrinsic factor impact on exposure (organ impairment, age, race, concomitant meds) and potential dose modifications 155156168164.
  • When warranted by clinical risk, hepatic impairment studies in cancer patients to inform dosing have been requested 379.

Why this matters: FDA expects a quantitative rationale for first-in-human and subsequent dosing that supports benefit–risk and anticipates labeling content; weak dose justification is a frequent gap that can delay IND progress 5130132.

Clinical trial design and endpoints

Common feedback emphasizes clarity and robustness of endpoints and analyses:

  • ORR and DOR by blinded independent central review (BICR) often underpin early efficacy signals; FDA has asked for side‑by‑side RECIST-based analyses, inclusion of all eligible patients (including those with non‑measurable disease), and alignment of the primary efficacy population with BICR-assessed measurability 48913445.
  • For accelerated approval, FDA stresses that response must be sufficiently large and durable, with confirmed responses and no detriment to OS; unconfirmed responses are not acceptable for ORR 234230.
  • Disease control rate (DCR) is not generally acceptable for labeling as a measure of clinical benefit compared with time‑to‑event endpoints; FDA has recommended using ORR (with prespecified multiplicity control) and/or PFS as primary endpoints depending on context 31391242241239.
  • Trials should pre-specify statistical analysis plans, including alpha allocation for multiple endpoints, interim futility criteria, and subgroup strategies with appropriate control of false positives 206356234.
  • Early incorporation of well-defined patient‑focused outcome measures (PRO/COA) is encouraged, with discussion prior to confirmatory trials 1982631673033.
  • Program-specific guidance appears frequently: pediatric LGG requests for RANO/RAPNO‑based analyses and sufficient follow‑up; advice that confirmatory trials be substantially enrolled for accelerated approvals 366482.

Why this matters: Endpoint selection and analysis rigor drive interpretability, supportability for accelerated pathways, and ultimately labeling claims; FDA often conditions IND advancement and registrational strategies on these design features 234242241.

Patient population, eligibility, and representativeness

FDA regularly probes who is studied and how that maps to U.S. practice:

  • Representativeness: IND-stage protocols should describe how the clinical trial population will reflect U.S. patients by disease characteristics, demographics, and standards of care, including participation across U.S. and non‑U.S. sites 403996529.
  • Eligibility and stratification: requests include clarifying inclusion/exclusion (e.g., anticoagulation in HEPZATO), adding biomarker or mutation‑based stratification (e.g., BRAF V600E vs V600K), and defining how biomarker positivity is established 4231756872.
  • Single‑arm trials: sponsors should justify why randomization is not feasible and plan adequate sample sizes to ensure precision around ORR (e.g., confidence interval width) 393199.

Why this matters: Credible generalizability and sound stratification enable the Agency to interpret efficacy and safety in U.S. practice and can be decisive for accelerated approval decisions 399175199.

Safety monitoring and analysis expectations

FDA feedback consistently addresses safety signal characterization and documentation:

  • Pooled analyses for AESIs (e.g., immune‑mediated events), standardized MedDRA queries, and pre‑specified safety issues with analytic strategies are commonly requested 388491373.
  • Narrative requirements (e.g., deaths within 30 days of last dose, SAEs, discontinuations) and safety update schedules are reiterated at IND and pre‑NDA junctures 362365256223.
  • Sponsors must follow expedited reporting timelines (7‑ and 15‑day reports) and maintain ongoing integrated safety summaries 230.

Why this matters: Robust, prespecified safety analyses and narratives facilitate early signal detection and later labeling; gaps here can stall IND amendments and complicate reviews 230388.

Data standards, submission logistics, and inspection readiness

Even at IND preparation, FDA stresses data quality and organization:

  • Use of CDISC‑compliant datasets (with ADRG/SDRG) and reviewer guides that reconcile legacy vs. standard datasets; early identification of lab units (U.S. conventional and SI) to avoid conversion issues 19838538166.
  • eCTD compliance and use of the ESG; OSI/BIMO packages in standardized electronic format to support inspection planning for pivotal trials 92280190192.
  • Clear cover letters for new/amended protocols with study phase, objectives, population, design, and anticipated issues to facilitate timely feedback 198.

Why this matters: INDs that are technically sound and inspection‑ready smooth the path to early advice and future NDA/BLA reviews; noncompliance can lead to delays or refusals to receive 192280.

CMC/manufacturing expectations

Recurrent CMC themes appear across biologics, small molecules, ADCs, and device–drug combinations:

  • Comprehensive, readily located lists of all clinical sites and manufacturing/testing facilities, including FEI numbers and roles, are expected in the application 219280274210168.
  • Stability packages at submission (e.g., long‑term and accelerated durations) and clarity on retest/shelf‑life; FDA may accept limited long‑term data at filing with final determinations during review 108104.
  • Starting materials designation and impurity fate/purge data, with FDA reserving the right to request upstream starting materials based on review 2426.
  • Control strategies and methods: resolution of clinically relevant in vitro release methods (when applicable), justification of NIR or non‑routine tests, dissolution method development and comparative dissolution expectations, and PQAA submission to support CMC review 265426521.
  • Validation approaches and process robustness, including allowance for concurrent validation with small batch sizes in justified cases and high‑level process validation plans/design space concepts aligned to ICH Q8/Q9/Q10 219541.
  • For device‑enabled regimens, FDA often requires resolution of device validation and human factors before trial initiation 80513.
  • Documentation quality: English translations of batch records when applicable; organized Module 3 with summaries rather than raw reports only 407406.

Why this matters: CMC completeness at IND and early in development de‑risks clinical supply and future inspection outcomes; incomplete site lists, weak stability packages, or unresolved method issues are frequent review bottlenecks 219108426521.

Pediatric and companion diagnostic planning

  • Pediatric Study Plans: For many oncology drugs, FDA expects an Initial Pediatric Study Plan (iPSP) within 60 days of End‑of‑Phase 2; lack of an agreed plan should not block filing, but plans must be submitted and justified 310170.
  • Biomarker/CDx alignment: For biomarker‑selected indications, FDA asks for gene‑level efficacy reporting, prespecified variant definitions, and alignment with an analytically validated test; in some cases, an IDE for the CDx is advised prior to pivotal trial initiation 7268494.
  • Tissue‑agnostic vs tumor‑specific: FDA may signal that current data are insufficient for tissue‑agnostic claims and advise focusing on tumor‑specific indications 13.

Why this matters: Early pediatric and CDx planning prevents downstream surprises and informs inclusion criteria, endpoint strategies, and labeling pathways 3107213.

Common requests for additional studies or analyses

While not universal, FDA frequently asks for targeted additional work when data are incomplete:

  • Clinical pharmacology: DDI studies (including transporter interactions), hepatic impairment, population PK/ER analyses, and robust QT/QTc characterization 9141742257256.
  • Nonclinical/toxicology: Completion of chronic tox and reproductive assessments in certain programs before marketing trials; auditing GLP compliance for foreign studies relied upon for U.S. submissions 86209.
  • Human factors: Inclusion of a human factors validation study where user risk is material (e.g., drug–device systems or complex administration) 21480.
  • Confirmatory strategy: For accelerated approvals, FDA often seeks evidence of an enrolling confirmatory trial and adequate follow‑up to characterize durability 369482.

Why this matters: These targeted asks are predictable in oncology and can be scheduled proactively in the development plan to avoid gating issues at IND or pre‑NDA milestones 41786369.

Use of FDA review pilot programs and process guidance

FDA regularly highlights process options and expectations that affect timelines:

  • Oncology Center of Excellence pilots such as Real‑Time Oncology Review (RTOR) and Assessment Aid to facilitate interactive and efficient reviews, with encouragement to notify FDA as topline data mature 190174533.
  • Clear expectations for rolling submissions (only complete sections are acceptable) and for including IDMC minutes, BICR charters, and safety updates at agreed intervals 533107104108.

Why this matters: Early alignment on process can materially compress review timelines and clarify documentation expectations that trace back to IND planning 174533.

Practical implications for sponsors in 2025

  • Arrive at IND with a defendable quantitative dose rationale and a clear plan to close known gaps (DDI, QT, impairment studies) aligned to labeling‑relevant analyses 1301325417.
  • Lock primary endpoints and analysis plans with multiplicity control; plan BICR and durability assessment if pursuing accelerated pathways; embed PROs/COAs early 48923424224119833.
  • Define eligibility, biomarker rules, and stratification up‑front; document U.S. representativeness and, for single‑arm designs, justify feasibility and ensure sample sizes provide precision around ORR 1757240393199.
  • Pre‑wire safety analytics (AESIs, narratives, pooled strategies) and expedited reporting processes; ensure inspection‑ready data standards and reviewer guides 388491362198190192.
  • Assemble full facility lists, stability packages, and control strategies; for device‑involved products, complete validation and human factors work streams before pivotal trials 21910842680.
  • Submit iPSPs on time and engage early with CDRH on CDx strategies where applicable; reconsider tissue‑agnostic claims unless broadly supported 3107213.

Taken together, these patterns represent the most common FDA feedback in Type B minutes for solid tumor therapies preparing to file an IND or planning early registrational strategies. Addressing them proactively can de‑risk IND acceptance and accelerate time to pivotal execution 130132234242198922802191084263107213.