Virtual Library

Start Your Search

Jacob Sands



Author of

  • +

    MA09 - EGFR & MET (ID 128)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Targeted Therapy
    • Presentations: 1
    • Now Available
    • +

      MA09.02 - In Vivo, Ex Vivo and Early Clinical Activity of EGFR Monoclonal Antibody and Osimertinib in EGFR Exon 20 Insertion NSCLC (Now Available) (ID 968)

      15:15 - 16:45  |  Author(s): Jacob Sands

      • Abstract
      • Presentation
      • Slides

      Background

      EGFR Exon 20 insertions (Ex20Ins) are the 3rd most common class of EGFR activating mutation, but patients with NSCLC harboring EGFR Ex20Ins lack effective approved EGFR-TKIs. Newer-generation TKIs and combination strategies with EGFR-monoclonal antibodies (moAbs) may enhance activity against EGFR Ex20Ins.

      Method

      Xenografts derived from CRISPR-modified H2073 cells with Ex20Ins (A763_Y764InsFQEA, D770_N771InsSVD or V769_D770InsASV) and Ex20Ins patient-derived xenografts (PDXs) (D770_N771InsSVD, A797_V769dupASV, D770_N771_InsG, H773_V774_InsNPH) were treated with vehicle, osimertinib , cetuximab, and osimertinib+cetuximab. Ex20Ins spheroid models (D770_N771InsSVD and M766_A767InsASV) were treated with cetuximab at fixed dose and increasing concentrations of osimertinib. Ex20Ins PDX (A763_Y764InsFQEA) was also treated with afatinib and erlotinib; Ex20Ins PDX (D770_N771InsSVD) was treated with these combinations plus afatinib+cetuximab. Immunoblotting for pharmacodynamic studies of on-target and downstream proteins, phospho-proteins and apoptosis markers were performed at relevant timepoints for D770_N771InsSVD PDX and CRISPR model. A phase 1 clinical trial with a dose expansion cohort in Stage IV EGFR Ex20Ins NSCLC is currently open to accrual at osimertinib 80 mg qd and the EGFR-moAb necitumumab 800 mg IV D1 and D8 of 21D cycle with response assessment by RECIST 1.1 (NCT02496663).

      Result

      The combination of osimertinib and cetuximab achieved significant tumor growth inhibition compared to osimertinib alone across PDX and CRISPR cell line xenograft models (p=0.05), except for the A763_Y764InsFQEA PDX model where osimertinib alone and osimertinib+cetuximab were equivalently effective (both p<0.001 compared to control). Spheroid models for D770_N771InsSVD and M766_A767InsASV showed significantly increased cytotoxicity from the addition of cetuximab across multiple doses of osimertinib. Osimertinib+cetuximab was superior to erlotinib, cetuximab, afatinib and afatinib+cetuximab in a D770_N771InsSVD PDX model (p<0.001). In this model, inhibition of p-EGFR, p-ERK, p-HER2 and increased caspase 3 cleavage were noted, consistent with significant tumor growth inhibition. In the phase 1 EGFR Ex20Ins expansion cohort of necitumumab in combination with osimertinib, 6/18 patients enrolled with 4 patients evaluable for response; 2 patients achieved a partial response and median PFS was 5.3 months.

      Conclusion

      In vivo and ex vivo modeling in CRISPR cell line xenografts, PDXs and organoids demonstrated preclinical activity of dual EGFR blockade with osimertinib and EGFR monoclonal antibody in the 5 most common EGFR Ex20Ins representing a frequency of ~60% of detectable EGFR Ex20Ins in clinical practice. Osimertinib alone was as active as osimertinib plus cetuximab in A763_Y764InsFQEA, consistent with known sensitivity of this proximal insertion to single-agent EGFR-TKI. In a phase 1 study, osimertinib and the EGFR moAb necitumumab demonstrates preliminary clinically activity in EGFR Ex20Ins NSCLC.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

  • +

    MA15 - Usage of Computer and Molecular Analysis in Treatment Selection and Disease Prognostication (ID 141)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Pathology
    • Presentations: 1
    • Now Available
    • +

      MA15.06 - Stage I Lung Adenocarcinoma Gene Expression Associated with Aggressive Histologic Features for Guiding Precision Surgery and Therapy (Now Available) (ID 1124)

      15:45 - 17:15  |  Author(s): Jacob Sands

      • Abstract
      • Presentation
      • Slides

      Background

      Stage I lung adenocarcinomas (LUADs) show heterogeneity in histologic patterns which correlate with malignant behavior. Solid, micropapillary and cribriform patterns are associated with worse survival whereas lepidic (in situ) predominance has the best prognosis. In this study, we sought to characterize histologic pattern specific gene expression in resected clinical stage I LUADs. We also aimed to train and validate a genomic biomarker predictive of histologic aggressive patterns with the ultimate goal of being able to impact surgical and therapeutic decision making for post-biopsy management.

      Method

      A training cohort of 56 tumors from patients meeting NCCN high-risk screening criteria with stage I LUAD was included for pathologic annotation and whole exome RNA sequencing. Histologic pattern subtyping in 5% increments including all diagnostic slides was performed. A single representative FFPE block was chosen for RNA library-prep with Illumina TruSeq Access Kit and sequencing. Negative binomial models were used to identify gene expression differences associated with percent solid, cribriform, or micropapillary histology, and EnrichR was used for gene pathway enrichment analysis. Ss-GSEA was used to predict tumor infiltration of 20 immune cell types. A random-forest classifier for predicting aggressive histologic patterns was trained using 5-fold cross validation. A set of tumors from 16 independent patients with ≤2.0 cm clinical stage I LUAD was macro-dissected into 32 paired components (lepidic + non-lepidic regions) and subjected to RNAseq. Six tumors were defined as non-aggressive (lepidic + acinar/papillary) and ten tumors were defined as aggressive (lepidic + solid/micropapillary/cribriform). Four aggressive tumors were upstaged after surgical resection.

      Result

      In the training cohort, we identified 1322 genes associated with tumor histologic composition(FDR q <0.05 and fold-change > 2). Genes whose expression differs with solid histology% are enriched for involvement in DNA replication, cell cycle regulation and inflammation (FDR q<0.001). Genes whose expression is associated with micropapillary% are enriched for involvement in tRNA-aminoacylation and decrease of T-cell activity (FDR q<0.001). The functional enrichment of genes whose expression is associated with cribiform% was less informative. LUADs with micropapillary patterns exhibited gene expression consistent with decreased antigen presentation and low T-cell infiltration, and solid patterns exhibited gene expression consistent with increased infiltration of T-regulatory and Th2 cells (FDR q<0.05).

      A gene expression classifier was trained to predict the presence of aggressive histologic patterns. We validated this classifier on a set of 16 tumor specimens from which we macro-dissected and analyzed tissue from the most aggressive histologic pattern (AUC = 1.00). We also found that this classifier could differentiate lepidic regions isolated from aggressive tumors from lepidic regions isolated from non-aggressive tumors (AUC = 0.74).

      Conclusion

      We identified solid-, micropapillary- and cribriform-specific gene expression and associated immune response among clinical stage I LUADs, and developed a classifier predictive of aggressive histologic features using either lepidic (in situ) or non-lepidic components. As such, this biomarker has the potential to predict histologic aggressiveness even from pre-surgical tumor biopsies where all histologic patterns may not be represented. Such a biomarker may be useful in guiding clinical decision making including extent of surgical resection.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

  • +

    MA25 - Precision Medicine in Advanced NSCLC (ID 352)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Advanced NSCLC
    • Presentations: 1
    • Now Available
    • +

      MA25.10 - First-In-Human Phase 1 Study of DS-1062a (TROP2 Antibody-Drug Conjugate) in Patients with Advanced Non-Small Cell Lung Cancer (Now Available) (ID 3854)

      14:30 - 16:00  |  Author(s): Jacob Sands

      • Abstract
      • Presentation
      • Slides

      Background

      DS-1062a is a trophoblast cell-surface antigen 2 (TROP2)-targeting antibody-drug conjugate with Daiichi-Sankyo exatecan derivative (DXd) technology. TROP2 is highly expressed in epithelial cancers, including non-small cell lung cancer (NSCLC), and is associated with poor survival. In preclinical studies DS-1062a showed promising antitumor activity in xenograft mouse models. Updated results from the dose escalation part of a phase 1 study of DS-1062a in patients with advanced NSCLC are reported.

      Method

      This is an ongoing US and Japan dose-escalation/dose-expansion phase 1 study of DS-1062a in patients with unselected NSCLC (NCT03401385). Adult (age ≥20 years [Japan] or ≥18 years [US]) patients with measurable disease per RECIST v1.1 and available tumor for TROP2 measurement were eligible. The primary objectives are to identify the maximum tolerated dose (MTD) and recommended dose for expansion, assess safety and tolerability. Endpoints include safety, efficacy, pharmacokinetics, and molecular and genomic analyses.

      Result

      At most recent data cutoff (April 12, 2019) 39 patients with advanced NSCLC were treated with DS-1062a at doses of 0.27 (n=4), 0.5 (n=5), 1.0 (n=7), 2.0 (n=6), 4.0 (n=6), 6.0 (n=8) and 8.0 (n=3) mg/kg. Overall, patients were exposed to a median (range) of 3.0 (1–10) treatment cycles over a duration of 8.86 (3.0–31.1) weeks. Patient disposition included dose interruption (n=2), reduction (n=1) and discontinuation (n=23; primary reason was progressive disease (PD) per RECIST in 13/23 patients). The majority (87.2%; 34/39) of patients reported ≥1 treatment-emergent adverse event (TEAE), regardless of severity or causality; the most common (in ≥30% of patients) were fatigue (33.3%) and nausea (30.8%). Grade ≥3 TEAEs were reported in 41.0% (16/39) of patients, of which 12.5% (2/16) were considered drug related. Drug-related TEAEs occurred in 59.0% (23/39 [21/23 grade 1 or 2], and serious TEAEs in 25.6% (10/39 [n=8 grade 3 (n=1 grade 5/sepsis/6.0-mg/kg dose; n=1 grade 3/drug-related/maculopapular rash/6.0-mg/kg dose; n=1 grade 2/drug-related/pyrexia/4.0-mg/kg dose) of patients. One DLT (maculopapular rash, grade 3; resolved) occurred with the 6.0-mg/kg dose; the MTD has not been reached. Of tumor-evaluable patients, as of May 23, 2019, 10 partial responses (PR) were observed (7 PRs were observed at the April 12, 2019 datacut), with a clear dose response and good durability: n=1 in the 2mg/kg, n=2 in the 4-mg/kg, n=3 in the 6 mg/kg, and n=4/5 evaluable in the 8.0-mg/kg groups (4 of the PRs remain to be confirmed). Across all dose groups (April 12, 2019 datacut), 16 stable disease (SD), and 11 PD were observed. Systemic DS-1062a exposure increased in an approximate dose-proportional manner; plasma DS-1062a levels and total anti-TROP2 antibody were similar, suggesting DS-1062a stability in circulation. Updated tumor response profile and durability, biomarker analyses and correlation with clinical outcome will be presented, including immunohistochemistry and circulating tumor DNA analysis of baseline and sequential on-treatment samples, and other related markers.

      Conclusion

      DS-1062a was well tolerated and 10 PRs were observed during dose selection in unselected NSCLC patients having progressed on standard of care, including immune checkpoint inhibition in 8 of 10 patients. Updated data will be presented.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

  • +

    PC01 - Reinventing Clinical Trials (ID 83)

    • Event: WCLC 2019
    • Type: Pro-Con Session
    • Track: Advocacy
    • Presentations: 1
    • Now Available
    • +

      PC01.01 - Setting the Stage: Tension Between Patient Safety, Scientific Purity and Patient Inclusion (Now Available) (ID 3555)

      11:00 - 12:30  |  Presenting Author(s): Jacob Sands

      • Abstract
      • Presentation
      • Slides

      Abstract

      Clinical trials are important to the process of better understanding the therapeutic potential of new drugs while also serving as treatment options for individuals seeking the best possible treatment. When writing protocols, we think about how best to determine the effectiveness and side effect profile of the treatment being studied. This can lead to more limited eligibility in an effort to reduce complications not related to the disease or drug. When seeing patients in clinic, we think about how best to treat each individual, which can lead to preferring less strict eligibility. These two perspectives can sometimes seem to be at odds. How do we best define eligibility in a way that both provides scientific rigor while also providing the trial as an option to as many people as possible? This session sets the stage for an in-depth discussion of these 2 important aspects of clinical trials and how to strike the ideal balance for enrollment eligibility criteria.

      As part of this introduction, a highlight of the process of drug development through different phases of clinical trials from “pre-clinical work” to phase 3 studies is helpful. The following is a general structure. Any prospective drug must demonstrate compelling results at one phase to then be studied in the next phase. This starts with lab studies and often treatment of mice with prospective drugs. Any studies before treatment of humans is called “pre-clinical.” When results in this setting are promising, treatment in humans starts with phase 1. In this setting the focus of the study is to find the most appropriate dose, and there are often multiple diagnoses allowed for enrollment within that one study. The strategy for dose finding generally includes starting treatment for a few individuals at a low starting dose. After verifying tolerability, another group starts treatment on the study at a higher dose and so on. Less commonly, studies will allow dose increases for each individual that is tolerating the lower dose with ongoing disease control. Dose escalation often continues until finding the “maximum tolerated dose.” Although the side effects of the drug often weigh heavily in determining the dose for further study, responses to the treatment are certainly considered. After determining a dose for future study, this drug may enter a phase 2 study. In this phase, many more individuals are treated at the determined dose, and the effectiveness of the treatment is studied within a specific clinical setting. Results that suggest efficacy often lead to a phase 3 study, which includes randomization to the new drug, or the standard of care treatment, within a specific clinical setting. Although placebo is sometimes utilized, all individuals should get at least the best known therapy. For example, KEYNOTE-189 enrolled individuals with metastatic nonsquamous, non-small cell lung cancer (without sensitizing EGFR or ALK mutations) to the standard of care first line chemotherapy, pemetrexed and a platinum, with randomization to also include either pembrolizumab or placebo. Although people received placebo, it was given along with the chemotherapy, as was pembrolizumab.

      This introduction is followed by two related session. One is focused on crafting trials to allow more broad enrollment with the acknowledgment of trials as a treatment option. The other is focused on trial development with full attention toward scientific rigor and study of the drug. A discussion of the balance of these goals follows.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.