Tagged: Research Review

Targeting Ras in Cancer Therapies: Advances in Protein Engineering

In a new review, researchers from The Hebrew University of Jerusalem discuss the challenges associated with targeting Ras proteins and how protein engineering has emerged as a promising method to overcome these challenges.

Figure 3: Various scaffolds utilized to engineer binders to Ras and their binding epitopes. Targeting Ras in Cancer Therapies: Advances in Protein Engineering
Figure 3: Various scaffolds utilized to engineer binders to Ras and their binding epitopes.

Ras plays a crucial role in controlling various cellular processes by switching between active (Ras-GTP) and inactive (Ras-GDP) states with the help of specific molecules. In its active form, Ras interacts with multiple effector proteins, initiating downstream events. Humans have three Ras genes, resulting in four isoforms that have distinct expression patterns and unique functions in different tissues. Posttranslational modifications target Ras to the cell membrane, where it can form dimers and interact with effectors through common domains. Ras mutations, commonly found in pancreatic, colorectal and lung cancers, lock Ras in an active state, promoting continuous cell division and proliferation. Ras signaling disruption occurs through reduced catalytic activity, altered effector binding and decreased affinity for other regulatory proteins.

Although Ras has been considered difficult to target, recent advancements have identified potential binding pockets that can be addressed by small molecules, peptidomimetics and proteins. Inhibitors designed to covalently bind to the Ras G12C mutant have shown promise, leading to FDA-approved drugs for specific lung cancers. Additionally, protein-based inhibitors that target Ras and its interactions with effectors, regulatory proteins and guanine nucleotide exchange factors offer alternative strategies for therapeutic intervention. These developments have challenged the notion that Ras is “undruggable” and highlight the potential for effective treatments against various cancer types.

On July 1, 2023, researchers Atilio Tomazini and Julia M. Shifman from The Hebrew University of Jerusalem published a new review paper in Oncotarget, entitled, “Targeting Ras with protein engineering.” The authors provide an overview of the challenges associated with targeting Ras proteins with small molecules and discuss how protein engineering has emerged as a promising method to overcome these challenges.

“While the development of small-molecule Ras inhibitors has been reviewed elsewhere [40], we focus our review on protein-based Ras inhibitors, describing the methods for their engineering, various scaffolds used for inhibitor design, and prospects for delivery of the designed Ras inhibitors into the cellular cytoplasm, where Ras is located.”

Protein Engineering

Protein scaffolds offer alternative approaches to small molecule drugs for engineering protein-based inhibitors. Unlike small molecules, protein domains can bind to targets through large surface areas, providing high affinity and specificity. Antibodies, natural protein effectors and novel binding domains are commonly used as protein scaffolds. Antibodies can be engineered into smaller versions to overcome limitations, while natural effectors can be modified to enhance binding affinity. Novel binding domains, unrelated to the target protein, possess structural robustness and can be evolved to exhibit strong binding. All three classes of protein scaffolds have been utilized to engineer Ras binders and explore strategies to inhibit Ras oncogenesis.

“Interestingly, all classes of protein scaffolds, including antibodies, natural effectors, and novel binding domains, have been utilized for engineering of Ras binders, allowing scientists to target various sites on the Ras surface and to explore different strategies for inhibiting Ras oncogenesis […].”

Methods for engineering protein inhibitors can be categorized into experimental directed evolution and computational design, or a combination of both. Experimental techniques involve display technologies such as phage display, yeast surface display, ribosome display, and mRNA display. These methods allow for the construction of combinatorial libraries of protein mutants, which are then screened using the target protein as a selection “bait.” The selected binders are sequenced to identify high-affinity mutants. Negative selection steps can be incorporated to enhance specificity by eliminating binders to unwanted targets. The number of mutants that can be assayed depends on the display technology used, with each approach having its limitations.

In addition to experimental approaches, computational methods have been proposed for protein binder design. Computational design enables rational targeting of specific binding epitopes on the target protein. However, computationally designed binders often have weak initial binding affinities and require affinity maturation through experimental techniques. Computational methods have been successful in designing focused libraries for yeast surface display experiments, where small libraries of protein mutants are designed based on computational predictions. This approach narrows down the choices to the most promising mutants, facilitating directed evolution experiments. By combining computational and experimental approaches, protein inhibitors with superior affinity and specificity have been developed.

“We have summarized all the described engineered Ras protein-based binders and their properties in Table 1.”

The Future of Intracellular Transport for Ras Inhibitors

Efficient delivery of molecules that bind to intracellular Ras proteins is essential for suppressing pro-cancer pathways and promoting anti-cancer activities. To overcome the challenge of crossing the cell membrane, different strategies have emerged. One approach involves utilizing short cell-penetrating peptides (CPPs) that can be fused to the desired protein, allowing entry into cells through direct translocation or endocytosis. However, improving the release of cargo proteins from endosomes remains a hurdle. Supercharging proteins with positively charged surfaces or leveraging bacterial toxins with intrinsic delivery mechanisms are alternative methods for intracellular protein delivery. Additionally, coupling cargo proteins to nanoparticles or employing mRNA delivery systems have shown promise, although they have their own limitations.

These protein delivery techniques have been explored for targeting Ras inhibitors. For instance, a human IgG1 antibody was engineered to selectively bind to Ras-GTP, inhibiting downstream signaling. Fusion of Ras binding domains to CPPs demonstrated competitive inhibition of Ras/effector interactions. Furthermore, optimized bacterial secretion systems and lipid nanoparticle-encapsulated mRNA platforms have been employed for efficient intracellular delivery of Ras-binding molecules. These advancements open up possibilities for targeted cancer therapies and disease treatments by enabling effective delivery of Ras binders to their intracellular target, thus influencing cancer-related signaling pathways.

Conclusions

In summary, targeting Ras proteins, despite their historically challenging nature, has seen significant progress in recent years. Small molecules, peptidomimetics and protein-based inhibitors have emerged as potential strategies for inhibiting Ras oncogenesis. Protein engineering, utilizing various protein scaffolds such as antibodies, natural effectors and novel binding domains, offers alternative approaches to traditional small molecule drugs.

Experimental directed evolution and computational design, alone or in combination, have facilitated the development of high-affinity and specific protein inhibitors. Furthermore, the efficient intracellular delivery methods described above hold promise for targeted cancer therapies by effectively delivering Ras binders to their intracellular targets. These advancements challenge the perception of Ras as “undruggable” and provide hope for the development of effective treatments for various cancer types.

“These strategies should be utilized in future to examine the beneficial activity of Ras-binders and inhibitors and should further facilitate the development of protein-based Ras therapeutics.”

Click here to read the full review in Oncotarget.

ONCOTARGET VIDEOS: YouTube | LabTube | Oncotarget.com

Oncotarget is an open-access, peer-reviewed journal that has published primarily oncology-focused research papers since 2010. These papers are available to readers (at no cost and free of subscription barriers) in a continuous publishing format at Oncotarget.com. Oncotarget is indexed/archived on MEDLINE / PMC / PubMed.

Click here to subscribe to Oncotarget publication updates.

For media inquiries, please contact media@impactjournals.com.

Trending with Impact: Review of HER2 Variants in Breast Cancer Tumors

This review compiles splice variations in HER2 breast cancer, specifically in the context of the tumor environment, and co-expression of variants. The study also provides an up-to-date (as of Nov. 2020) account of HER2 and HER2 variant patterns of resistance to anti-HER2 therapies and other interventions.

Photomicrograph of immunohistochemistry for HER2, showing positive cell membrane staining in this infiltrating ductal carcinoma
Photomicrograph of immunohistochemistry for HER2, showing positive cell membrane staining in this infiltrating ductal carcinoma

The Trending with Impact series highlights Oncotarget publications attracting higher visibility among readers around the world online, in the news, and on social media—beyond normal readership levels. Look for future science news and articles about the latest trending publications here, and at Oncotarget.com.

According to Cancer Research UK, breast cancer occurs in one in every eight women within their lifetime and is the second highest cause of cancer related deaths in the UK. Breast cancer is a blanketed term for a wide variety of tumors that occur in the mammary glands. In over 20% of breast cancers, the human epidermal growth factor receptor two gene, officially named ErbB2 but otherwise known as HER2, is overexpressed. HER2 is a member of the epidermal growth factor receptor family of EGFR, HER2, HER3, and HER4. Overexpression of the HER2 protein was discovered in 1987 as a biomarker associated with poor prognosis and aggressive tumor types in breast cancer. This finding has accelerated research studies and progress in HER2 diagnostic testing and targeted therapeutics. However, the issue of HER2 resistance in these targeted therapies remains problematic.

“At the present time, we have an incomplete understanding of why patients with HER2+ breast cancer exhibit variable responses or resistance to targeted therapies [7374].”

Researchers from the Translational and Clinical Research Institute at Newcastle University in the United Kingdom have compiled a review of variations in HER2 breast cancer, specifically in the context of the tumor environment and when multiple variants are co-expressed at altered ratios. Their study also provides an up-to-date (as of Nov. 2020) account of the current landscape of HER2 variants and links this to patterns of resistance against HER2 therapies and other interventions.

“It is clear HER2 expression is not as simple as a single oncogenic overexpressed protein. It is likely many variants, arising from splicing and other mechanisms, are present in tandem. The relative ratios of these are likely to fluctuate depending on cellular conditions, during tumorigenesis and breast cancer progression.”

HER2 Variants & Co-expression

This paper provides an exquisitely detailed description and explanation of the HER2 protein structure, signaling pathways, sub-typing, and in-depth treatment functionality of a number of different HER2 targeted therapeutics. 

“Different forms of the HER2 protein exist within tumours in tandem and can display altered biological activities.” 

The unique interest in researching variations in HER2 breast cancer has increased since the identification of Δ16-HER2: a particular splice variant and link to resistance of anti-HER2 therapies. The “Δ16” in Δ16-HER2 refers to the lack of exon-16, which encodes a small extracellular portion of the DNA. Δ16-HER2 represents approximately 9% of the normal HER2 transcripts and its expression is considered common in breast cancer. Previous studies have identified Δ16-HER2 and HER2 normal transcripts can be co-expressed at varying levels in breast carcinomas. 

In the variant P100, less is known about this truncated HER2 protein. It has been hypothesized that P100 reduces the efficacy of monoclonal antibody HER2 treatments.

The splice variant Herstatin is produced by the retention of intron-8 in the HER2 protein. Herstatin acts as a tumor suppressor by blocking HER2 activity and cell proliferation, while promoting apoptosis. The researchers mention that it is important to note that cells expressing high levels of Herstatin are more sensitive to Tamoxifen.

“It’s noteworthy that one study proposed that the presence of Herstatin transcript does not segregate by tumour grade or size, patient age, lymph node involvement or ER status and that mRNA transcripts were present in matched non-cancerous breast tissue and breast carcinomas [96].”

Researchers in this review note that identifying and assessing the expression ratios of these different variants and classifying them as prognostic and predictive biomarkers may aid in further personalized treatment of breast cancer in HER2 positive patients.

Testing and Research Landscape

“Studying splicing regulation and how this is altered in breast cancer could explain patterns of expression and how these link to treatment resistance [111].”

The researchers write that tests assessing for both HER2 status and HER2 variant expression could potentially refine their predictions of a patients’ response to treatment. One common way that researchers gauge levels of HER2 proteins, and only some HER2 variants, is through immunohistochemistry tests. mRNA assessments are also used to identify gene expression patterns. Another biomarker test the researchers noted that may be best used for prognostic predictions is the Enzyme-Immunoassay—to assess levels of plasma or serum HER2 (sHER2) in the blood produced by cleavage or splicing.

“Cohort studies have identified sHER2 testing as a useful complementary test to IHC owing to the correlations between high sHER2 and aggressive tumour phenotypes such as invasion and metastases.”

Targeted HER2 Treatments

The review elaborates in detail about targeted treatments for HER2 breast cancer, which include: trastuzumab, pertuzumab, lapatinib, and T-DM1. They note that endocrine therapy is utilized for ER positive patients and chemotherapy, radiotherapy, and surgery are all still utilized.

Conclusion

“Work in vitro and in vivo as well as analysis from clinical trials has identified patterns of resistance to the standard of care treatment options in HER2+ patients which are correlated to variant expression.” 

This goal of their review was to summarise the current landscape of HER2 variant research and to explain why researchers should consider HER2 variant levels and ratios when offering the best treatment plan for breast cancer patients.

Click here to read the full review, published in Oncotarget.

Oncotarget is a unique platform designed to house scientific studies in a journal format that is available for anyone to read—without a paywall making access more difficult. This means information that has the potential to benefit our societies from the inside out can be shared with friends, neighbors, colleagues and other researchers, far and wide.


For media inquiries, please contact media@impactjournals.com.