Tag: cancer

Performing target validation well

Performing target validation well

Summary

This blogpost describes issues encountered in target validation and how to safeguard against poor reproducibility in RNAi experiments.

The importance of target validation

More than half of all clinical trials fail from a lack of drug efficacy. One of the major reasons for this is inadequate target validation.

Target validation involves verifying whether a target (protein/nucleic acid) merits the development of a drug (small molecule/biologic) for therapeutic application.

Failing to adequately validate a target can burden a pharma with roughly 800 million to 1.4 billion in drug development costs. Impact is not only monetary as large site closures  often result as companies struggle to save costs and a reduced production effort deprives patients of new medicines.

Performing target validation well

Special attention should therefore be given to performing target validation techniques well.

target validation techniques
Overview of target validation techniques (Lindsay, Nat Review Drug Discovery, 2013)

Many of these techniques involve inhibiting target expression to establish its relevance in a cellular or animal disease model. This can be performed with chemical probes, RNA interference (RNAi), genetic knock-outs, and even targeted protein degradation.

The reproducibility of these techniques however has been an issue of concern for drug developers. Less than half of all findings from peer-reviewed scientific publications was reported to be successfully reproduced.

Dismal rates of reproducibility from several pharma-led cancer-focused studies ranged from 11% (Amgen) to 25% (Bayer). A review by William Kaelin Jr sums up the common pitfalls of preclinical cancer target validation. One of his key points:

Cellular phenotypes caused by a chemical or genetic perturbant should be considered to be off-target until proved otherwise, especially when the phenotypes were detected in a down assay and therefore could reflect a nonspecific loss of cellular fitness. It is only by performing rescue experiments that one can formally address whether the effects of a perturbant are on-target.

The comment highlights the issue of reagent non-specificity as a notable contribution towards poor reproducibility.

Certainly, for RNAi the wide-spread off-target effects of siRNAs has been observed in numerous publications. The mechanism being well-established to be based on microRNA-like seed-based recognition of non-target genes. The effect dominates over on-target effects in many large RNAi screens, illustrating the depth of the problem.

Reagent non-specificity is not restricted to RNAi. There have been multiple reports of non-specificity for gene editing technique, CRISPR, which can be read about in detail here, here and here. Recent publications continue to shed more light on its potential off-targets as we learn more about this relatively new technique.

Even chemical probes may have multiple targets. It is hence imperative that more than one target validation technique be used to avoid confirmation bias.

Target validation – a story from Pharma

Back in 2013, when siTOOLs was just starting out, a pharma approached us with a target validation problem.

They were obtaining different results with 3 different siRNAs in a cellular proliferation assay. Despite all 3 siRNAs potently downregulating the target gene, they produced different effects on cell viability.

Which siRNA tool to trust?

target validation siRNA vs siPOOL pharma story
Three different siRNAs against the same target were tested in a cell proliferation assay. Despite all 3 siRNAs showing potent target gene silencing, effect on cell proliferation differed greatly.

A whole-transcriptome expression analysis performed for the 3 siRNAs and a siPOOL designed against the same target revealed the reason for the large variability.

target validation expression analysis siRNA vs siPOOL pharma story
How many genes can you affect with an siRNA? Whole transcriptome analysis by microarray was performed and number and % of up and down-regulated genes are shown over total number of genes assayed (18567).

Despite all siRNA tools affecting the same target, the difference in extent of gene deregulation was astounding. With the greatest number of off-target effects, it was not surprising that siRNA 3 showed an impact on cell proliferation.

In contrast, siPOOLs had 5 to 25X less differentially expressed genes compared to the 3 commercial siRNAs against the same target. An expression analysis carried out for another gene target showed similar results i.e. siPOOLs having far less off-targets.

The target was dropped from development. A great example where failing early is a good thing, though it was not without costs from validating the multiple siRNAs.

The recommended target validation tool

Functioning like a pack of wolves, siPOOLs increase the chances of capturing large and difficult prey, while making full use of group diversity to compensate for individual weakness.

siPOOLs efficiently counter RNAi off-target effects by high complexity pooling of sequence-defined siRNAs. This enables individual siRNAs to be administered at much lower concentrations, below the threshold for stimulating significant off-target gene deregulation. Due to having multiple siRNAs against the same target gene, target gene knock-down is maintained and in fact becomes more efficient.

siRNA vs siPOOL rtqPCR knock-down efficiency
siPOOLs increase targeting efficiency, avoiding knock-down variability. Figure shows rtqPCR quantification of target RNA levels when two siPOOLs vs two siRNAs against 36 genes were tested.

We still recommend using multiple target validation techniques. As a first evaluation however, siPOOLs are quick, easy and most of all, reliable.

Rescue experiments can also be performed with siPOOL-resistant rescue constructs.

Find out more

Novel anti-cancer mechanism identified by shRNA/siRNA off-target effects

Novel anti-cancer mechanism identified by shRNA/siRNA off-target effects

Summary:

siRNA off-target effects takes an interesting turn for cancer research as reported in eLIFE by Putzbach et. al. Research unveiled a specific group of survival genes in cancer cells thanks to the off-target effects of siRNAs/shRNAs.

death receptor signaling pathways
CD95 highlighted in death receptor signaling pathways

 

CD95 is a death receptor that mediates apoptosis when bound to its ligand, CD95L or FasL. Known for its multiple tumour-promoting activities, it was not surprising that silencing both molecules by RNAi produced cancer cell death.

What was surprising –  death induced by C95/CD95L siRNAs/shRNAs did not work through CD95/CD95L at all. Three observations contributed to this conclusion:

  1. The toxicity correlated with siRNA/shRNA concentration

Using siRNAs at 0.1nM or expressing the shRNA in a miR-30 backbone (developed to reduce off-targets by expressing shRNAs at reduced levels) did not induce the same toxicity

 

  1. Removing the target did not affect siRNA/shRNA-induced toxicity

Excising the siRNA/shRNA binding sites on CD95/CD95L with CRISPR did not protect cells from toxicity induced by these siRNAs/shRNAs

 

  1. Restoring expression of the target did not rescue cells from siRNA/shRNA-induced toxicity

Introducing recombinant CD95/CD95L proteins or expressing siRNA-resistant versions of CD95/95L, did not rescue cells from toxicity induced by their siRNA/shRNAs

 

The case of shRNA/siRNA off-target effects

The evidence was pretty convincing that the toxic effects of the CD95/CD95L siRNA/shRNAs stemmed from off-target effects.

1. Step-wise mutations showed toxicity derived from the seed sequence 

siRNA sequence step-wise mutation shows siRNA off-target effects
siRNA sequence step-wise mutation

Substituting each base of the tox-inducing siRNA (siL3) with the non-toxic siRNA (siScr) sequence in a step-wise cumulative manner either from the seed end or the non-seed end, highlighted toxicity derived from the seed sequence. The seed sequence is a 6-base sequence at position 2 to 7 of the guide RNA strand and is responsible for defining the off-target profile of an siRNA (read this technote for more information)

2. Off-target survival genes identified by RNA-seq

An RNA-seq analysis of CD95 or CD95L shRNA-treated cells identified twelve genes with significantly altered expression levels – 11 downregulated, 1 upregulated:

Death induced by survival gene elimination identified by siRNA off-target effects
Genes regulated by toxic shRNA were important survival genes

 

It turns out that many of the downregulated genes were important for survival. Additionally, two recent genome-wide lethality screens independently identified six of these genes (highlighted in red). The authors therefore termed this form of CD95/CD95L siRNA/shRNA-induced cell death Death Induced by Survival Gene Elimination (DISE). Don’t we all love acronyms! As depicted, these genes mostly interfered with apoptosis, cell cycle, autophagy and senescence.

3. Survival genes targeted by miRNA-like activity of CD95/CD95L siRNA/shRNAs

Sylamer plots and seed matches to survival genes showing siRNA off-target effects
Survival genes were enriched for seed matches at the 3′ UTR to toxic shRNA

The seed sequence is what microRNAs (miRNAs) use to recognize and downregulate target genes. siRNAs/shRNAs can behave like miRNAs, contributing to the off-target activity. As shown by Sylamer plots, the seed sequence of toxic shRNAs (shL3 and shR6) were enriched in highly downregulated genes. The identified survival genes also contained multiple seed matches over their 3’ UTRs. That leaves little doubt that the CD95/CD95L shRNAs were hitting these genes through miRNA-like off-target activity.

Conclusion:

Once again, we see how siRNA off-target effects can impact experimental results. Though in this case, it actually helped identify relevant survival genes! Notably, siRNA off-target effects likely influence cell viability/proliferation data to a greater extent than other readouts since it is regulated by so many genes.

This is not an isolated report of siRNA off-targeting in cancer. Targets such as STK33 and MELK, identified with RNAi to be important in cancer progression, failed to show the same effects in experiments performed by different groups or alternative techniques. The controversy continues however as their effects on cancer activity continue to be reported.

How to avoid siRNA off-target effects

siPOOLs were developed to avoid siRNA off-target effects through high complexity pooling and optimized design. Phenotypes are therefore more clearly and reliably ascribed to loss-of-function of the target gene. The new siPOOL Cancer Toolbox now provides cancer researchers the ability to disrupt multiple genes reliably, with reduced risk of siRNA off-targets, in an affordable toolkit solution.

siTOOL top 100 cancer gene list for siPOOL Cancer Toolbox

We scoured the published literature for the most highly cited genes involved in multiple forms of cancer. Choose your target genes from our list of top 100 cancer-associated genes to build your own siPOOL Cancer Toolbox.  Notably, CD95, MELK and STK33 did not make the cut!

See our top 100 cancer gene list

CRISPR/Cas9 Screening – The “Copy-Number Effect”

CRISPR/Cas9 Screening – The “Copy-Number Effect”

Several CRISPR/Cas9 screens identifying essential genes in cancer cell lines have been performed to date (Shalem et al., 2014, Hart et al., 2015, Kiessling et al., 2016). These typically take the form of pooled screens where sgRNA libraries targeting all genes or subsets of genes are introduced in parallel into Cas9-expressing cells, at a single sgRNA per cell. The sgRNAs exert a negative or positive selection pressure on cells based on their impact on cell viability and proliferation. The most depleted or enriched sgRNA sequences are determined by next-generation sequencing, revealing relevant gene ‘hits’. Very similar to how pooled shRNA screens are performed.

From these screens, several groups have observed a worrying phenomenon: CRISPR gRNAs targeting genomic regions of high copy number amplification showed a striking reduction in cell proliferation/survival. Dr William Hahn’s group at the Dana Farber Institute was one of the first to characterize this in a publication last year involving a CRISPR/Cas9 screen on 33 cancer cell lines looking for essential genes. In total, 123411 unique sgRNAs were used targeting 19050 genes (6 sgRNAs/gene), 1864 miRNAs and 1000 non-targeting negative control sgRNAs.

What they discovered is a little worrying to say the least.

The figure shows two genomic regions in two different cell lines (SU86.86 and HT29). At genomic coordinates highlighted by the red box, 3 tracks are shown. Top, copy number from the Cancer Cell Line Encyclopaedia (CCLE) SNP arrays, red indicating above average ploidy and blue showing below; middle, CRISPR/Cas9 guide scores with purple trend line indicating the mean CRISPR guide score for each CN segment defined from the above track; bottom, RNAi gene-dependency scores. AKT2 and MYC, known driver oncogenes at these loci, respectively, are highlighted in orange. For RNAi data, shRNAs targeting AKT2 used in Project Achilles were not effective in suppressing AKT2 (hence the negative result).

 

Key findings:

  • A striking enrichment of negative CRISPR guide scores (i.e. sgRNAs that reduced cell proliferation/survival) for genes that reside in genomic regions of high copy-number amplification.

 

  • Genes identified in CRISPR that reduced survival, did not have the same effect when disrupted by RNAi in the same cell lines (this RNAi screen was done by the same group but published 2 years before).

 

  • This enrichment was seen also for unexpressed genes, i.e. genes not transcribed. Meaning the reduced survival was not due to loss-of-function of the targeted gene.

 

  • Even for regions with low absolute copy numbers, a significant reduction in survival was observed compared to non-targeting control sgRNAs. Furthermore, the effect was dose-dependent with greater copy number amplifications producing larger negative CRISPR guide scores.

Notably, the correlation between copy number and genes that were scored high on essentiality was also observed when looking at data from other studies (Hart et al., 2015). The “copy number effect” would therefore produce a high number of false positives in CRISPR screens for essential genes in cancer cell lines. The graph above shows just how big an effect this is. Comparing genes identified as essential in a CRISPR screen vs RNAi screen, increasingly essential CRISPR-identified genes were more likely to reside on copy number amplifications (defined as having average sample ploidy > 2). This effect was notably absent for RNAi-derived essential genes.

Aside from false positives, the increased noise due to “copy number effects” also increases false negatives. MET, a gene identified by shRNA screens, for example, failed to be picked out by CRISPR screens as it is located on a chromosome 7 amplicon (7q31) in MKN45 cells (gastric cancer cell line) where all other gRNAs within that amplicon also scored as essential.

The authors go on to explore mechanisms behind the “copy number effect”. They found it was attributed to a DNA damage response stimulated by excessive cutting by Cas9. This response appeared p53-dependent and induced cell cycle arrest at the G2 phase, explaining the anti-proliferative effect. A similar response was seen for promiscuous sgRNAs that cut at multiple sites, with effects being more pronounced when cuts were spread over several chromosomes as opposed to a single chromosome.

How to manage this?

So far, most simply avoid analysing hits where sgRNAs lie at amplified regions or target multiple sites (Wang et al., 2017). However, these regions of copy number amplifications have been implicated in cancer and may contain relevant hits. Several computational methods have therefore recently been developed to correct for “the copy number effect”. Hahn’s group developed a computational algorithm called CERES based on data obtained from CRISPR sgRNA screens in 342 cancer cell lines representing 27 cell lineages.

Novartis also developed a Local Drop Out (LDO) algorithm that corrects obtained data based on examining gRNAs scores at direct genomic neighbours. When multiple neighbouring genes show similar drop out scores, effects are assumed to be due to “copy number effects”. This method has the advantage of not requiring prior knowledge of copy number, however it does require a sufficient density of gRNAs to accurately capture “copy number effects”.  They also had an alternative method, Generalized Additive Model (GAM) where copy number was taken into account.

 

How the CERES Model Works

The Results – copy number dependency is reduced while preserving essentiality of cancer-specific genes such as KRAS

 

A step towards the right direction but the penetrance of this effect still raises some concerns:

  • Although false positives are reduced with these computational methods, it is difficult to recapture false negatives. This is dependent on the gRNA having a stronger phenotype compared to neighbouring gRNAs on the amplicon which is not always the case. The LDO method for example still failed to recapture MET.

 

  • Guide scores can vary with cell line, sgRNA and experimental conditions, making it difficult to apply the same counter-measures to every experiment.

 

  • Given multiple cut sites trigger the same effect, how do we ensure multiple sgRNAs when introduced into a cell are not inducing a similar response? This is difficult to control in pooled screens, and poses a limitation in multiplex screens. Synthetic lethality screens for example with sgRNAs targeting multiple genes, might be subject to a higher false positive rate.

 

  • With even diploid genes (copy number = 2) having statistically significant growth reduction compared to haploid gene loci, the challenge still remains to delineate a true loss-of-function over a non-specific cellular response.

 

  • Negative sgRNA controls have to be carefully selected. From the study, non-targeting controls had little impact on viability compared to most other sgRNAs. Controls targeting non-expressed genes or non-essential loci have been recommended as better controls.

 

  • Lastly, although this effect seems to apply mostly to cancer cell lines that undergo a high rate of gene amplifications, similar effects may extend to polyploid tissues such as the liver.

Hence as always gene function should be determined by a variety of methods. Using RNAi for example to affirm a CRISPR-knockout phenotype would add greater confidence to a hit. To avoid those RNAi-related false positives however, its probably best to use siPOOLs.

 

Source of figures:

Aguirre, A. J., Meyers, R. M., Weir, B. A., Vazquez, F., Zhang, C.-Z., Ben-David, U., … Hahn, W. C. (2016). Genomic Copy Number Dictates a Gene-Independent Cell Response to CRISPR/Cas9 Targeting. Cancer Discovery, 6(8), 914 LP-929.

Meyers, R. M., Bryan, J. G., McFarland, J. M., Weir, B. A., Sizemore, A. E., Xu, H., … Tsherniak, A. (2017). Computational correction of copy-number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells. bioRxiv. Retrieved from https://biorxiv.org/content/early/2017/07/10/160861.abstract

Other relevant sources:

Munoz, D. M., Cassiani, P. J., Li, L., Billy, E., Korn, J. M., Jones, M. D., … Schlabach, M. R. (2016). CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions. Cancer Discovery, 6(8), 900 LP-913. Retrieved from https://cancerdiscovery.aacrjournals.org/content/6/8/900.abstract

de Weck, A., Golji, J., Jones, M. D., Korn, J. M., Billy, E., McDonald, E. R., … Kauffmann, A. (2017). Correction of copy number induced false positives in CRISPR screens. bioRxiv. Retrieved from https://biorxiv.org/content/early/2017/06/23/151985.abstract

 

Want to receive regular blog updates? Sign up for our siTOOLs Newsletter:

Loading
error

Like what you see? Mouse over icons to Follow / Share