Very excited to share our synthetic lethality based precision oncology approach via the tumor transcriptome (SELECT) @Cell
This effort is led by @joo_sang_lee @theNCI #PrecisionMedicine [1/n]
cell.com/cell/fulltext/…
Current precision oncology mainly targets actionable mutations & has limited patient coverage. We present the first precision oncology framework that systematically guides cancer treatment based on *synthetic lethal* vulnerabilities identified via the tumor transcriptome.
*Synthetic lethality* (SL) is an interaction between a pair of genes, where their individual inactivation has no phenotype, but their co-inactivation leads to cell death. We identified the SL pairs from analyzing a large collection of patient cohorts of 10k tumors.
The basic idea underlying the use of SLs for guiding patients is that a drug is likely to be more effective in patients where many of the SL partners of its targets are under-expressed in the tumor, thus leading to specific and selective tumor cell death.
SELECT prediction accuracy was tested on a large collection of 35 clinical trial datasets (spanning 14 targeted therapy and 21 immunotherapy cohorts across 10 cancer types), a scale markedly surpassing previous efforts to our knowledge.
For targeted therapy, SELECT is predictive of patients’ tumor response in 7 out of 10 clinical trial datasets with accuracy levels of AUC of ROC>0.7 and successfully predicts patient survival in 3 out of 4 additional cohorts.
For checkpoint immunotherapy, SELECT is predictive of tumor response in 15 out of 18 clinical trial datasets with accuracy levels of AUC>0.7 and successfully predicts patient survival in 3 additional cohorts (Gide et al. also shows AUC>0.7 in addition to the survival signal).
This collection includes a fresh pre-treatment transcriptomics profiles and treatment response data from Samsung Medical Center, where lung adenocarcinoma patients were treated with pembrolizumab (ncbi.nlm.nih.gov/geo/query/acc.…).
Overall, through a retrospective analysis of 35 targeted or immunotherapy clinical trials of about 2,500 patients, SELECT is predictive of response in 28 out of 35 cases (80%) across 10 different cancer types.
Evaluating this approach retrospectively in a recent WINTHER multi-arm basket clinical trial, we show that SELECT is predictive of the response and that the fraction of patients benefitting from treatments may possibly be markedly increased by targeting SL vulnerabilities.
The attached figure illustrates reports that a clinician would receive from a SELECT analysis. Based on pre-treatment transcriptomic profiles of patients, an SL-score will be assigned to each drug, providing clinicians with personalized SL-based treatment options.
SELECT is the first synthetic lethality-based precision oncology tool for analyzing the tumor transcriptome. It shows considerable accuracy across many different targeted and immunotherapy trials, laying a basis for future prospective transcriptomics-based clinical trials.
Without the dedication of clinicians and patients involved in all the trials analyzed in this study, this research wouldn’t have been possible. We greatly thank them and made our codes and datasets publicly available for academic purposes (zenodo.org/record/4661265).
Many thanks to our co-authors @NishanthUNair @KunWang331 @Sanjusinha7 and others, and thanks to our wonderful collaborators @NCIKenAldape @Dr_R_Kurzrock @MRGneuroonc @hannenhalli @ZeevRonai and many more!! Read the full paper @CellCellPress :)

• • •

Missing some Tweet in this thread? You can try to force a refresh
 

Keep Current with Eytan Ruppin, M.D., Ph.D.

Eytan Ruppin, M.D., Ph.D. Profile picture

Stay in touch and get notified when new unrolls are available from this author!

Read all threads

This Thread may be Removed Anytime!

PDF

Twitter may remove this content at anytime! Save it as PDF for later use!

Try unrolling a thread yourself!

how to unroll video
  1. Follow @ThreadReaderApp to mention us!

  2. From a Twitter thread mention us with a keyword "unroll"
@threadreaderapp unroll

Practice here first or read more on our help page!

More from @NCIEytanRuppin

3 Apr
Can we infer an individual’s tissue-specific expression from their whole blood transcriptome?
Hot off the Press & a summary of our effort is below!
advances.sciencemag.org/content/7/14/e…
with @hannenhalli & @KunWang331
Learning an individual’s tissue-specific gene expression can be invaluable guiding diagnosis & monitoring progression of a wide range of diseases. Unfortunately, for most tissues, the transcriptome cannot be obtained without invasive procedures.
In contrast, measuring whole blood expression is minimally invasive and high time resolution.
Read 8 tweets
16 May 20
Excited to share our preprint introducing the first pipeline to identify Cell type Specific Intracellular (CSI) microbes from single cell RNA-seq data. Here's a tweetorial summary. biorxiv.org/content/10.110…
Multiple recent studies have pointed to the functional importance of tumor microbiome. One eg., is the case of Fusobacterium in primary & metastatic colon tumors, which drives tumorigenesis, influences response to chemotherapy & binds to multiple human immune inhibitory receptors
Among many, the key challenges to systematically chart tumor microbiome are: 1. Remove reads due to contamination, 2. Whether microbes are intra/extra cellular and 3. if intra, which cell types within the tumor do they reside in?
Read 11 tweets
1 Apr 20
We present (preprint) an approach for identifying drug treatments modulating the expression of a key protein required for the SARS-CoV-2 virus entry – the ACE2 (angiotensin-converting enzyme 2) receptor. preprints.org/manuscript/202…
Two weeks ago, Fang et al suggested that diabetes and hypertension patients treated with ACE-inhibitors & AR-blockers are at a higher risk for COVID-19 as these treatments may increase ACE2 expression.
This motivated us to ask this question – Which clinically approved drugs including antihypertensives may modulate ACE2 in lung tissue?
Read 10 tweets
23 Feb 20
In a promising step for cancer precision medicine, we devised a novel strategy that identifies genetic interactions and robustly predicts drug response, spanning 21 targeted therapies and 11 immunotherapy datasets across 10 different cancer types (1/9) bit.ly/38Pzn9m
To predict treatment outcomes, we defined genetic interaction scores, which capture the likelihood of a given patient responding to a given drug. (2/9)
We predicted treatment response in 17 of the 21 targeted- and 8 of the 11 immuno- therapy datasets, with considerable accuracy (AUC > 0.7). (3/9)
Read 9 tweets
18 Feb 20
We present the first computational strategy leveraging genetic interactions successfully predicting patients response to many different targeted and immunotherapy treatments based on their tumors transcriptome. Sharing the details here- biorxiv.org/content/10.110… Image
Current precision oncology is mainly performed by targeting actionable mutations & has a limited patient coverage. Here we present the first approach for systematically guiding patients treatment based on vulnerabilities identified across the whole exome.
We stratified responders in 17 out of 21 different targeted therapies cohorts with an AUC>0.7 across different cancer types better than previously published transcriptome-based signatures. Image
Read 11 tweets
3 Nov 19
We identified for the first time a selection of three cancer driver mutations (P53, KRAS and VHL) during CRISPR-Cas9 gene editing and chart a comprehensive gene-wise editing risk map in our new work out today. This series summarizes it 1/X biorxiv.org/content/biorxi…
Recent reports in @NatureMedicine suggested that CRISPR-Cas9 gene editing induces a p53-dependent DNA damage response in primary cells, which may select for cells with oncogenic p53 mutations. These CRISPR-induced possible changes needs a systematic and thorough testing. 2/X Image
Leveraging computational and experimental techniques we asked whether CRISPR gene editing may select for other oncogenic mutations and if yes, which genes editing could severely induce this selection. 3/X Image
Read 10 tweets

Did Thread Reader help you today?

Support us! We are indie developers!


This site is made by just two indie developers on a laptop doing marketing, support and development! Read more about the story.

Become a Premium Member ($3/month or $30/year) and get exclusive features!

Become Premium

Too expensive? Make a small donation by buying us coffee ($5) or help with server cost ($10)

Donate via Paypal Become our Patreon

Thank you for your support!

Follow Us on Twitter!