Non-small cell lung cancer (NSCLC)

The most common type of lung cancer

Epigenetic Alterations in Blood as Markers for Early Lung Cancer Detection

Early Detection Research Award
Grant title (if any)
Rising Tide Foundation for Clinical Cancer Research/LUNGevity Foundation Lung Cancer Early Detection Award
This grant was co-funded by Rising Tide Foundation for Clinical Cancer Research
Abhijit Patel, MD, PhD
Yale University
New Haven
CT
Steven Skates, PhD
Harvard Medical School
Cambridge
MA

The objective of this project is to develop a blood test that can improve upon current limitations in lung cancer screening.  Dr. Patel and his team have developed a method to accurately measure alterations in DNA that are cancer-specific by looking at levels of methylation of circulating tumor DNA (ctDNA) in the bloodstream.  Using this method, Dr. Patel will develop a predictive model to identify patients with lung cancer based on these DNA alterations at a single time point, as well as an algorithm that can track these changes in a patient’s DNA over time.  If successful, this could help detect lung cancer earlier in its development, thereby leading to better outcomes for patients.

Research Summary

Lung cancer is by far the most deadly cancer in the U.S., with total lung cancer deaths exceeding those of the next three major cancers combined. Such dismal statistics are largely attributable to the insidious nature of the disease; by the time symptoms appear, the cancer has often spread to an extent that makes cure unlikely or impossible. In contrast, patients who are diagnosed at earlier stages have much better outcomes, as their tumors can be entirely removed or eradicated prior to distant spread. Thus, annual chest CT scans for lung cancer screening have proven to be effective at reducing lung cancer deaths, and are currently recommended for patients with a heavy smoking history. However, CT-based screening programs have been practically challenging to implement, and uptake has been slow. An alternative screening approach that has been garnering much enthusiasm is based on development of a simple blood test that detects DNA fragments shed from tumor cells into the bloodstream. Several commercial and academic groups have been racing to develop blood tests for cancer screening based on this concept, and the field has made impressive progress. However, detection of early-stage lung cancers has remained particularly challenging, with sensitivities reaching only ~20-40% for Stage I disease. A key limitation for detection of small, early-stage tumors has been the extremely low abundance of DNA fragments bearing cancer-specific features (such as mutations) in the circulation. To overcome this limitation, our group has developed a technology that can accurately measure cancer-specific alterations in DNA which are more highly abundant (known as “hypermethylation”). In the current project, we propose to develop a predictive model to identify patients with lung cancer based on probabilities inferred from measurement of these DNA alterations. We will then further improve the sensitivity for detecting the earliest stages of lung cancer by developing an algorithm that tracks longitudinal changes in a patient’s DNA signal over time rather than relying on just a single time-point.

Technical Abstract

Early detection of cancer has long been one of the grand challenges of medicine. It is widely acknowledged that better methods for detection of small, asymptomatic tumors are likely to translate to substantial improvements in cancer survival rates. This is an especially important priority for lung cancer because of its high incidence, high rate of late-stage diagnosis, and high mortality. Over the past decade, liquid biopsy approaches based on detection of cancer-specific mutations or epigenetic changes in cell-free DNA (cfDNA) have made significant inroads towards this goal. However, detection of early-stage lung cancer has been particularly challenging because of the minute amounts of tumor DNA shed into blood. Methylation of cfDNA has emerged as a biomarker of choice for many early detection efforts, but existing technologies are designed to probe for cancer-specific methylation patterns either at pre-specified target sites or across broad genomic regions. The former approach prioritizes a limited subset of cancer-relevant signals, whereas the latter approach yields sparse cancer signals from extensive sequence data. Our group has developed a liquid biopsy technology that comprehensively profiles hypermethylated promoter sequences in cfDNA arising from anywhere in the genome. Using a high-stringency capture strategy based on methylation density rather than sequence, our method is able to globally profile hypermethylated promoters without pre-specifying targets. Gene silencing via promoter hypermethylation is a fundamental mechanism of carcinogenesis, and this aberrant signal can be detected at very low levels in plasma because background methylation patterns in healthy plasma are remarkably consistent. To optimize sensitivity for detection of early-stage lung cancer, we will develop a scoring scheme based on probabilistic machine learning to predict the likelihood of lung cancer by integrating hypermethylation signals across thousands of cell-free DNA fragments. Unlike most current liquid biopsy-based early detection efforts which are focused on identifying individuals with cancer based on a single time-point measurement, here we propose to develop a longitudinal early detection algorithm based on measurement of serial increases in cancer-specific epigenetic signals over time due to tumor growth and accumulating changes in the epigenome.

Novel Protein Degraders for Treating RET Positive Cancer

Partner Awards
Grant title (if any)
RETpositive/LUNGevity Lung Cancer Research Award Program
Justin Drake, PhD
University of Minnesota
Minneapolis
MN

This project will investigate novel protein degraders (called PROTACs) as a treatment for RET-positive cancers, and will evaluate their efficacy in vitro and in vivo in prostate and lung cancer. PROTACs are highly specific molecules that degrade unwanted or harmful proteins in cells (in this case, RET tyrosine kinase). This research aims to provide a novel therapeutic approach targeting RET signaling, which could overcome resistance to existing RET inhibitors.  If successful, it would be a first-in-class compound for further clinical development.

Research Summary

RET receptor tyrosine kinase is a proto-oncogene that requires a co-receptor and secreted ligand for activation. Activating RET mutations are oncogenic targets in non-small cell lung cancer, medullary thyroid cancer and neuroendocrine type cancers. Two RET-specific kinase inhibitors (BLU-667 and LOXO-292) have been FDA approved for treating RET fusion positive cancers. This has led to a tremendous advance in lung cancer therapy and objective response rates in patients naïve to RET inhibitors or who have received other RET inhibitors such as cabozantinib or vandetanib. It is also apparent that RET therapy-driven resistance is common and new alternatives are needed to drug this pathway. Our hypothesis is that targeting RET with novel, first-in-class RET degraders will result in cell death that is more durable than existing inhibitors of RET kinase activity. We will test these novel degraders on several models of prostate and lung cancer to assess on target RET degradation and efficacy in cell line and mouse models. Once novel RET protein degraders are developed and tested in prostate and lung cancer models in vitro and in vivo, we plan to perform pre-clinical optimization studies of compounds and work towards clinical implementation in late stage prostate, lung, and other cancers that rely on RET signaling for survival.

Technical Abstract

Two RET-specific kinase inhibitors (BLU-667 and LOXO-292) have been FDA approved for treating RET fusion positive cancers. This has led to a tremendous advance in lung cancer therapy and objective response rates in patients naïve to RET inhibitors or who have received other RET inhibitors such as cabozantinib or vandetanib. It is also apparent that RET therapy-driven resistance is common and new alternatives are needed to drug this pathway. Our hypothesis is that targeting RET with novel, first-in-class RET degraders will result in cell death that is more durable than existing inhibitors of RET kinase activity. We will test hypothesis via the following specific aims: Aim 1. Development and characterization of RET degraders for treating RET positive cancers and Aim 2. Evaluate efficacy of RET degraders in in vitro and in vivo NEPC models. In aim 1, we propose to develop RET degraders based on a new RET inhibitor, vepafestinib, and assess RET degrader specificity and activity using a panel of prostate and lung cancer cell line models that overexpress RET or contain RET fusions. We will then evaluate the efficacy of our RET degrader in in vitro and in vivo models of lung and prostate cancer. Once these novel RET protein degraders are developed and tested, we plan to perform pre-clinical optimization studies of compounds and work to identify a suitable pharma partner to develop for clinical implementation in late stage prostate, lung, and other cancers that rely on RET signaling for survival.

Developing new therapeutic approaches for RET-positive cancers

Partner Awards
Grant title (if any)
The Hamoui Foundation/LUNGevity Lung Cancer Research Award Program
Romel Somwar, PhD
Memorial Sloan Kettering Cancer Center
New York
NY

This project aims to develop new therapeutic approaches for RET-positive cancers, focusing on overcoming resistance to currently available RET inhibitors.  Dr. Somwar and colleagues will investigate ways to block the growth of lung cancers with altered RET in a pathway called MAPK (mitogen activated kinase), which is involved in many biological processes involving cell growth and survival.  MAPK is implicated in developing resistance to RET inhibitors and finding strategies to target this pathway in combination with RET could benefit many patients who have no approved therapy options after tumor reoccurence. 

Research Summary

Lung cancers are one of the leading causes of death in the US. Significant progress has been made over the past three decades to understand the biology of lung cancers and to stratify these diseases into subsets of patients who will get the maximum benefit of a given form of therapy. New technologies now allow for each patient to have their tumor DNA sequenced to find genetic causes of their cancer. Many genes that regulate cell growth are altered by mutations that cause the unrestricted growth that lead to cancer. Scientists have developed strategies to take advantage of these aberrant genes by finding chemicals or biological agents that will antagonize the protein products of these genes. One gene that is altered in 2% of lung cancers is called RET and there are two drugs that block the tumorigenic function of this cancer-causing gene (oncogene). Although patients respond very well to these two anti-RET drugs at first, they soon become resistant to the therapeutic effects. Additional genetic changes in RET or other genes in the cancer cells that regulate growth are responsible for the drug resistance. Our goal in this grant proposal is to find ways to block the growth of lung cancers with altered RET that stopped responding to anti-RET inhibitors. The strategy that we will test involves the simultaneous inhibition of RET and other proteins in another growth promoting pathway called the MAPK (mitogen activated kinase) pathway. We believe that this therapeutic strategy can benefit more than 30% of patients who stop responding to current drugs that target lung cancers with RET genetic alterations.

Technical Abstract

RET fusions result from abnormal rearrangements of the kinase domain of RET with other non-essential genes and drive tumorigenesis. These oncogenic chimeric tyrosine kinases are found in approximately  2% of non-small cell lung cancer (NSCLC).Two FDA-approved selective RET inhibitors (selpercatinib and pralsetinib) have shown great response rates in lung cancer patients. However, resistance to RET inhibitors inevitably occurs, limiting therapeutic benefit. Multiple mechanisms of resistance to RET inhibitors have been described, including acquired RET solvent front mutations (G810R/S/C/V), and RET-independent mechanisms of resistance due to amplifications of other receptor tyrosine kinases (RTK) including MET, FGFR1 and ERBB2, and alterations in the RAS-MAPK pathway. Some second-generation RET inhibitors that target secondary RET mutations have been recently developed including vepafestinib (TAS0953/HM06) which is currently being tested in phase I/II clinical trials in the US and Japan for RET fusion positive lung cancer. There is a clinical need to identify mechanisms of resistance to vepafestinib and develop strategies to overcome them.
 

RET with solvent front mutations, amplification of MET/FGFR1/ERBB2 and RAS-MAPK pathway mutations account for >30% of all resistance mechanisms to first-generation RET drugs, and importantly, all of these alterations are expected to activate the RASMAPK pathway. Therefore, a therapeutic strategy that tackles RAS-MAPK pathway activation is expected to benefit >30% of patients who acquire resistance to first-generation RET drugs. Moreover, given that RET fusions, like all tumors arising from activated RTKs engage the RAS-MPAK pathway for oncogenesis, we believe that many treatment-naïve patients may also benefit from a therapeutic strategy that targets RET and the RAS-MAPK pathway.  

Our first goal in this proposal is to simultaneously address resistance due to RAS-MAPK pathway alterations and extending the benefit of first-generation RET drugs by developing a combination therapy strategy involving RET and pan-RAS, MEK1/2 or ERK1/2 inhibitors. Our second goal is to decipher mechanisms by which the transcription factor capicua (CIC) regulate RET-driven tumorigenesis and resistance to RET inhibitors. We will perform transcriptomic, epigenic and proteomic profiling to gain insights into RET-ERK-CIC interaction. Our third goal is to identify and target resistance mechanisms to vepafestinib, so that a therapeutic strategy will be in place for when patients being treated with this drug develop resistance.

Our team includes leaders in the field of lung cancer clinical and translation research who have been at the forefront of lung cancer genomics and therapy, developing state of the art therapeutic strategies. We are well positioned to translate the findings from this study to the clinic within two years. These studies have the potential to benefit more than 30% of lung cancer patients with RET fusions.

Immunogenic peptide priming of dendritic cells for RET+ NSCLC

Partner Awards
Grant title (if any)
The Hamoui Foundation/LUNGevity Lung Cancer Research Award Program
Amy Cummings, MD, PhD
University of California, Los Angeles
Los Angeles
CA

This project will explore the use of neoantigens to evaluate immunogenic priming of dendritic cells (DC) in RET+ NSCLC.  Neoantigens are short protein fragments present only in cancer cells that bind to genetically encoded proteins known as human leukocyte antigens (HLA).  Dr. Cummings will use features of HLA to predict which cancer-specific protein fragments best match an individual’s immune system, utilizing a biobank of RET-rearranged NSCLC biospecimens. This approach could help identify optimal immunogenic targets, that could be translated into a pathway for clinical use of personalized DC vaccines.

Research Summary

RET-rearranged non-small cell lung cancer (NSCLC) is a rare subtype of lung cancer that is driven by growth signals triggered by RET activation. RET-specific inhibitors are effective initially, but most benefit from this treatment for only 1-2 years before additional treatment is needed. Chemotherapy is a widely-available option but typically provides less than six months of benefit, and it is unclear whether immunotherapy alone or in combination with chemotherapy is a better option. Findings from other gene-rearranged NSCLC studies, particularly those on ALK-rearranged NSCLC, suggest that immunotherapy works better when the immune system is better exposed to abnormalities created by the gene-rearrangement. These are neoantigens, or short protein fragments present only in cancer cells that bind to human leukocyte antigen (HLA), a scaffold that displays these protein fragments to the immune system. One issue with this approach is that these fragments have to be specifically matched to the immune system of an individual, and even the most common forms of HLA are only found in 20% of people. This means that these types of approaches would be applicable to at most 1 out of 5 people with RET-rearranged NSCLC. Our techniques broaden this approach by using features of HLA to predict which cancer-specific protein fragments best match an individual’s immune system (motif neoepitopes), including neoantigens from RET rearrangements and those predicted from the individual’s tumor. We propose to use our biobank of RET-rearranged NSCLC biospecimens, which have not been previously analyzed, to determine whether we can detect and elicit enhanced immune responses with motif neoepitopes, neoantigens related to RET-rearrangements, or other predicted neoantigens. We can then offer this approach in a currently open clinical trial investigating immune system optimization through an application to the FDA.

Technical Abstract

RET-rearranged non-small cell lung cancer (NSCLC) presents challenges in management following progression on selective tyrosine kinase inhibitors (TKIs). Platinum-based chemotherapy and docetaxel are available options but are without durable benefit. Real world data with single-agent and combination chemo-immunotherapy suggests modest benefit and possible efficacy if immunotherapy-based approaches are appropriately optimized. For the past decade, our group has meticulously curated hundreds of NSCLC biospecimens including matched tissue and blood from multiple timepoints, including 7 RET-rearranged NSCLC cases that have not previously been analyzed. We have extensive expertise in neoepitope prediction and personalized immunotherapy through dendritic cell (DC)-based vaccination. Our most recent collaboration enabled functional assessments of T-cells through nanovial-based affinity repertoires, further enhancing our ability to predict and translate immunogenic peptides through a personalized vaccine-based program. We propose to use our RET-rearranged NSCLC biospecimens to systematically study T-cell-specific responses to identify optimal immunogenic peptide targets, an approach that could be translated in our currently open and approved DC vaccination trial (NCT03546361) through single patient exemptions.

Targeting tumor associated macrophages in immunotherapy resistant NSCLC

Partner Awards
Grant title (if any)
Brown/LUNGevity Award to Understand Mechanisms of Resistance to Immunotherapy
Dwight Owen, MD, MSc
The Ohio State University
Columbus
OH

This project will investigate the role of cells called macrophages, key components of the immune system that have multiple functions, including immune surveillance within a unique communication pathway called hedgehog (Hh). The hedgehog signaling pathway is involved in cell growth and differentiation, as well as maintenance of stem cells and tissue repair. Disruption or inhibition of Hh can create an environment that is less favorable for survival of cancer cells, allowing a patient’s immune system to combat it more effectively.  This research has the potential to benefit patients who have been diagnosed with NSCLC, who have not responded to current treatments including immunotherapy by boosting the body’s own defense mechanisms.

Research Summary

Lung cancer remains one of the most lethal types of cancer worldwide, with non-small cell lung cancer (NSCLC) accounting for a majority of cases. The goal of our research is to better understand the relationship between certain immune cells called macrophages and NSCLC, and how this interaction contributes to the cancer's survival and resistance to treatment. The scientific premise of our project lies in investigating a unique communication pathway known as hedgehog signaling within these macrophages and determining how it impacts the immune system's ability to fight lung cancer. If successful, our research has the potential to benefit patients who have been diagnosed with NSCLC, particularly those who have not responded to current treatments including treatment with immune therapies. By disrupting the hedgehog signaling pathway in macrophages, we hope to create a tumor immune environment that is less favorable for cancer cell survival, allowing patients' immune systems to effectively combat the disease. This research can pave the way for innovative therapeutic approaches that boost the body's own defense mechanisms.

Technical Abstract

The prognosis for patients with metastatic non-small cell lung cancer (NSCLC) remains poor despite recent progress in immune checkpoint blockade (ICB) therapy. Thus, there is an urgent need to understand mechanisms for lung cancer immune evasion within the tumor microenvironment (TME) in order to develop more effective and durable strategies for treating lung cancer. Tumor associated macrophages (TAMs), a major component of the tumor stromal mass, generally display an anti-inflammatory phenotype and can facilitate tumor growth by promoting angiogenesis, invasion, and metastasis, as well as immune evasion. However, it remains largely undefined exactly how these TAMs regulate anti-tumor immune responses within the TME. Will test the hypothesis that hedgehog signaling in TAMs interferes with recruitment of CD8+ T cells to the TME through the following specific aims: 1) Investigate the role of Hh inhibition with anti-PD-L1 therapy in non-small cell lung cancer; 2) Study the impact of Hh inhibition on TAMs and changes within the TME. The objective of this project is to understand signals required for functional polarization of TAMs within the TME and its contributions to immune cell dysregulation and cancer progression, and whether combined Hh inhibition and ICB can overcome resistance to immunotherapy.

Developing EGFRxHER3 bispecific CAR-T cells for targeting EGFR TKI DTPCs

Career Development Award
Yan Yang, PhD
MD Anderson Cancer Center
Houston
TX

In patients with EGFR-mutant NSCLC, tyrosine kinase inhibitors (TKIs) have been an effective treatment, but over time these patients develop resistance to TKIs, leading to tumor relapse.  Dr. Yang’s project focuses on cancer cells called drug-tolerant persisters (DTPs), which are implicated in TKI resistance.  A gene called HER3 is expressed in DTPs, and Dr. Yang will use specially engineered immune cells, called CAR-T cells, to target both HER3 and EGFR simultaneously.  If successful, this approach would result in a bi-specific CAR-T cell that can be further evaluated in clinical trials.

Research Summary

In lung cancer patients with a specific genetic mutation in EGFR, certain drugs can be helpful, but over time, the cancer can learn to resist these drugs, making them less effective. We're focusing on a small group of tough cancer cells, called DTPCs, that survive the initial treatment because they might be the key to curing the cancer for good. We've noticed that a gene called HER3 is expressed more in these cells. We also have promising results from CAR-T cell therapy. It is a treatment using T cells, a type of immune cells, engineered with the chimeric antigen receptor (CAR) so they can find and destroy cancer cells. We've found that CAR-T cells targeting EGFR can kill DTPCs. We believe if we use these T cells to attack both HER3 and EGFR at the same time, we might have a better chance of killing off these stubborn cancer cells. To make sure this idea works, we'll first check if HER3 is indeed more expressed in these DTPCs in the lab and samples from patients. Then, we'll test our HER3-targeting CAR-T cells to see if they can kill these DTPCs. If that looks promising, we'll tweak the T cells to target both HER3 and EGFR and see if they work even better. If all goes well, we'll be able to make one of the first effective CAR-T cells to target both HER3 and EGFR and try them out in clinical trials.

Technical Abstract

In NSCLC patients harboring mutant EGFR, treatment with tyrosine kinase inhibitors (TKIs) has demonstrated efficacy. However, the emergence of resistance to EGFR TKIs remains a significant challenge, leading to disease progression. Targeting drug-tolerant persister cells (DTPCs), a rare subpopulation surviving initial treatment, emerges as a more effective strategy than awaiting the development of complete drug resistance, holding potential for curative cancer therapy. Based on our preliminary data on the upregulation of HER3 in DTPCs resistant to EGFR TKIs and the potent antitumor activity of EGFR-targeting chimeric antigen receptor (CAR)-T cells against DTPCs. We propose that HER3 is a promising target expressed on EGFR-TKI DTPCs and that CAR-T cells simultaneously targeting both EGFR and HER3 are an effective approach for targeting DTPCs with improved overall efficacy. In this proposal, we will evaluate HER3 as a therapeutic target for CAR-T cell therapy against EGFR-TKI DTPCs by validating its expression in DTPCs using in vitro cell models, in vivo xenograft and PDX models, and patient tissues. Additionally, we will evaluate the anti-tumor activity of HER3-targeting CAR-T cells against DTPCs. Subsequently, we will develop and characterize EGFRxHER3 bispecific CAR-T cells, evaluating their CAR expression, antigen binding affinity, and anti-tumor activity. We next will assess their anti-tumor efficacy in xenograft and PDX models to guide the selection of the most promising bispecific CAR-T cells for further development. If completed successfully, we will have EGFRxHER3 bispecific CAR-T cells ready for GMP-compliant clinical-grade CAR manufacturing, in preparation for clinical trial evaluation.

Building Reliable Oncology Navigation to Ensure Adjuvant Management: BRONx-TEAM Project

Career Development Award
Tamar Nobel, MD, MPH
Montefiore Medical Center
Bronx
NY

The introduction of targeted therapies and immunotherapy for early-stage lung cancer is associated with improved survival, but patients can only benefit if they partake in adjuvant and neoadjuvant therapies.  Data has shown that inequalities exist for patients with lower socioeconomic status as well as non-White patients when it comes to being referred for and receiving treatment after surgery.  These inequalities are likely to increase as new drugs are developed in clinical trials comprised of predominantly white patients.  In this project, Dr. Nobel will study the impact of disparities on uptake of adjuvant therapy for NSCLC in a largely minority patient population at Montefiore Medical Center in Bronx, NY.  She will provide social support and health literacy to engage patients in their care and collect genetic data about their tumors, which will contribute to future clinical trials that are more inclusive.

Research Summary

Systemic therapy after surgery to remove lung cancer has been demonstrated to improve survival. However, data has shown that there are inequalities in which patients are referred for and receive treatment after surgery, specifically for lower socioeconomic status and non-White patients. As new treatments have been developed, these inequalities are likely to increase as these drugs have been developed in clinical trials predominantly composed of White patients and the benefits in other populations are not known. We have previously demonstrated that using nursing and peer navigators to help guide patients in their cancer care improves treatment adherence in our predominantly Black and Hispanic low socioeconomic status population in the Bronx. The BRONx-TEAM project aims to improve patient outcomes by using a navigation pathway focused on increasing patient adherence to systemic therapy after surgery for non-small cell lung cancer resection. We believe that by providing social support and improving health literacy we can get patients to be more informed and engaged in their cancer care. Furthermore, we will gather genetic data about the patients tumors. Given our patient population, we have a unique opportunity to contribute to the literature to understand the relationships between tumor genetics, treatment types and outcomes in non-White patients. Furthermore, we will investigate the use of a commercial genetic panel to assess risk for recurrence. Given the lack of this type of data in low income non-White patients, we believe that this exploratory portion of our study will serve as an important foundation for future clinical trials that are more inclusive than the currently available literature.

Technical Abstract

As seen in the phase III trial CheckMate 816 (CM816), neoadjuvant anti-PD-1+chemotherapy improves survival for patients with resectable non-small cell lung cancer (NSCLC), with pathologic response as a major trial endpoint. Our team led the Central Pathology Review for CM816, and we showed the first prospective evidence that the full spectrum of % residual viable tumor (%RVT) associates with event free survival. Given the data supporting pathologic response as a survival surrogate, %RVT will likely be incorporated into the next generation of clinical trials and may ultimately guide clinical decision-making. %RVT is primarily evaluated using visual assessment of routine hematoxylin and eosin-stained slides. We developed a machine learning-based approach to score %RVT, which allows for a standardized approach that can be completed rapidly for a large volume of patients, and we propose to test this algorithm in resection specimens from CM816. Additionally, we will use multiplex immunofluorescence (mIF) to quantify individual features of pathologic response, locate them within the larger tumor bed, and determine the relative contribution in predicting patient outcomes. Furthermore, we will use the novel AstroPath platform, a mIF whole-slide imaging platform that uses algorithms first developed in astronomy to generate tumor-immune maps, to identify additional pre- and on-treatment biomarkers of response. Our goal is to leverage emerging technologies (i.e, machine learning and mIF) to develop the next generation of pathology biomarkers, including pathologic response assessment, and to identify additional features that can potentially be targeted in combination with anti-PD-(L)1+chemotherapy to improve clinical benefit in patients with NSCLC.

Next-generation pathologic response assessment in patients with lung cancer

Career Development Award
Julie Deutsch, MD
Johns Hopkins School of Medicine
Baltimore
MD

Dr. Deutsch’s proposal centers around finding better pathologic predictors of response to neoadjuvant IO in early stage NSCLC.  She will utilize machine learning/artificial intelligence to test an algorithm that she and her team have developed that assesses percent residual viable tumor (%RVT), which is the amount of tumor left at the time of surgery.  Dr. Deutsch will also characterize tissue specimens using a novel immunofluorescence platform to identify cell types and spatial relationships that are associated with patient benefit to immunotherapy+chemotherapy.  This approach can help inform which patients should receive a given therapy, how they will respond, and additional possible targets for the development of new therapies.

Research Summary

Immunotherapy revolutionized the treatment of lung cancer, and is now being extended so patients can receive therapy before surgery. This was supported by a large clinical trial, CheckMate 816 (CM816), where patients with lung cancer showed improved survival when treated with immunotherapy+chemotherapy before surgery, compared to chemotherapy alone followed by surgery. However, there is an unmet need to identify who is most likely to benefit from such an approach. To address this gap, we will apply novel, next-generation pathology biomarkers utilizing machine learning/artificial intelligence and multispectral imaging. Specifically, we have shown that the amount of tumor left at the time of surgery, termed percent residual viable tumor (%RVT), predicts survival. To date, %RVT assessment is primarily performed visually on glass slides using a light microscope. We developed a machine learning-based algorithm for assessing %RVT on digitized glass slides using a small cohort of patients at Johns Hopkins to improve standardization and throughput in preparation for broad usage. Here, we will test the algorithm’s performance in a larger cohort of patients (the CM816 patients). Additionally, we will characterize tissue specimens using the novel multiplex immunofluorescence AstroPath platform, which uses algorithms first developed in astronomy, to identify cell types and spatial relationships that are associated with patient benefit to immunotherapy+chemotherapy. Our goal is to use cutting-edge technologies to improve the care of lung cancer patients by informing which patients should receive a given therapy, how well patients will do after receiving therapy, and possible additional targets for the development of new therapies.

Technical Abstract

As seen in the phase III trial CheckMate 816 (CM816), neoadjuvant anti-PD-1+chemotherapy improves survival for patients with resectable non-small cell lung cancer (NSCLC), with pathologic response as a major trial endpoint. Our team led the Central Pathology Review for CM816, and we showed the first prospective evidence that the full spectrum of % residual viable tumor (%RVT) associates with event free survival. Given the data supporting pathologic response as a survival surrogate, %RVT will likely be incorporated into the next generation of clinical trials and may ultimately guide clinical decision-making. %RVT is primarily evaluated using visual assessment of routine hematoxylin and eosin-stained slides. We developed a machine learning-based approach to score %RVT, which allows for a standardized approach that can be completed rapidly for a large volume of patients, and we propose to test this algorithm in resection specimens from CM816. Additionally, we will use multiplex immunofluorescence (mIF) to quantify individual features of pathologic response, locate them within the larger tumor bed, and determine the relative contribution in predicting patient outcomes. Furthermore, we will use the novel AstroPath platform, a mIF whole-slide imaging platform that uses algorithms first developed in astronomy to generate tumor-immune maps, to identify additional pre- and on-treatment biomarkers of response. Our goal is to leverage emerging technologies (i.e, machine learning and mIF) to develop the next generation of pathology biomarkers, including pathologic response assessment, and to identify additional features that can potentially be targeted in combination with anti-PD-(L)1+chemotherapy to improve clinical benefit in patients with NSCLC.

Early detection and prognosis of lung cancer using bioengineered implants

Pierre Massion Young Investigator Award for Early Detection Research
Ramon Ocadiz Ruiz, PhD
University of Michigan
Ann Arbor
MI

Dr. Ocadiz Ruiz proposes to develop a bioengineered scaffolding and test it in mouse models.  If successful, this research could progress to a phase 1 clinical trial and lay the groundwork for a new technology to be used in individuals with increased risk of lung cancer. This technology has to potential to make biopsies and consequently, early detection, easier.

Comparative Effectiveness of Lung Cancer Screening Strategies

Pierre Massion Young Investigator Award for Early Detection Research
Lawrence Benjamin, MD
University of California Los Angeles
Los Angeles
CA

Dr. Benjamin’s research focuses on improving the rates of lung cancer screening. Currently, there is interest in “centralizing” lung cancer screening into self-contained programs or one-stop shops, with dedicated support staff and clinical personnel to coordinate shared decision-making, scheduling imaging, and arranging appropriate follow-up care. However, it is poorly understood how these centralized programs compare to “decentralized” screening that is coordinated by primary care physicians directly with their patients. Dr. Benjamin seeks to utilize nationwide longitudinal data from multiple lung cancer screening programs from the Veterans Affairs Healthcare System to evaluate and compare the performance of centralized versus decentralized screening programs, with particular focus on highlighting their effectiveness within various racial and income groups.