Therapeutic targeting of cell transition: ready for clinical prime-time? (2024)

  • Journal List
  • Cardiovasc Res
  • PMC11060484

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Therapeutic targeting of cell transition: ready for clinical prime-time? (1)

Link to Publisher's site

Cardiovasc Res. 2024 Apr; 120(5): e14–e16.

Published online 2024 Feb 16. doi:10.1093/cvr/cvae029

PMCID: PMC11060484

PMID: 38366667

Emiel P C van der VorstTherapeutic targeting of cell transition: ready for clinical prime-time? (2) and Jason C Kovacic

Author information Article notes Copyright and License information PMC Disclaimer

Associated Data

Data Availability Statement

Atherosclerosis, characterized by the build-up of lipid-laden plaques, is the main underlying cause of various cardiovascular diseases (CVDs), including myocardial infarction, peripheral vascular disease, arterial calcification, ischaemic cardiomyopathy, and a significant proportion of strokes. Endothelial cells, lining the luminal surface of the vessels, are major regulators of vascular hom*oeostasis and thereby play a crucial role in the development of atherosclerosis. Activation of endothelial cells by atherosclerotic risk factors results in cellular dysfunction and in the expression of pro-inflammatory cytokines and adhesion molecules, driving leukocyte migration and promoting the accumulation of leukocytes in the intima of the artery.

Over the last two decades, numerous studies have demonstrated that endothelial cells can acquire mesenchymal cell-like properties in a process called endothelial-to-mesenchymal transition (EndMT). During this transition, endothelial cells lose expression of classical cell markers, like VE-Cadherin and CD31, while they gain expression of mesenchymal markers, like alpha-smooth muscle actin. Through these cellular changes, EndMT contributes to CVDs like atherosclerosis1,2 and to other processes and diseases including the vascular remodelling and neointima formation following vein grant transplantation.3 Although the exact role and underlying mechanisms of EndMT in atherosclerosis and other CVDs are being rigorously investigated at the moment, so far there still remain large knowledge gaps.

The endothelium is a specialized type of epithelium. Just as endothelial cells can undergo EndMT, epithelial cells can also undergo epithelial-to-mesenchymal transition (EMT). As it happens, there is a far larger body of knowledge relating to EMT than EndMT, with EMT first formally described in the 1960s.4,5 Therefore, many lessons about EndMT can potentially be learned from the EMT process. Physiologically, EMT plays an important role in embryogenesis and wound healing, but on a pathological level, it has already been well established that EMT contributes to malignant progression, for example by increasing metastatic potential.6 Since carcinomas originate in epithelial tissues, EMT is understood to play a particularly important role in this type of cancer. The likely mechanism(s) whereby EMT promotes malignancy of epithelial tumours is by the acquisition of hallmark features of mesenchymal cells including increased motility, loss of cell–cell adhesion, and detachment from the epithelial basem*nt membrane. Due to its important role in carcinoma progression, it has been proposed that the inhibition of EMT may be a promising therapeutic oncological approach, although due to several limitations, concrete evidence for this clinical potential has remained elusive so far.

In a recent publication in Nature, Cassier et al.7 in their study Netrin-1 blockade inhibits tumour growth and EMT features in endometrial cancer show clear pre-clinical and clinical evidence that EMT inhibition indeed has great clinical potential. Focusing on endometrial adenocarcinomas, the authors were able to show that netrin-1 and its main receptor UNC5B are up-regulated in tumour tissue compared with normal endometrium. Based on this, Cassier et al. developed a monoclonal antibody called NP137 that neutralizes netrin-1 and blocks the netrin-1–UNC5B interaction. Treatment of mice with NP137 significantly decreased the development of endometrial tumours and increased the survival rate of these mice, demonstrating its potency. The authors then moved to study NP137 in patients with advanced endometrial carcinoma. In a Phase 1 trial with 14 patients who received NP137 every 2 weeks, there was no dose-limiting toxicity and 8 out of 14 patients achieved disease control (stable disease). Furthermore, at least in one exemplary case, NP137 treatment resulted in a striking 51% reduction of liver lesions within 6 weeks (Figure ​Figure11).

Open in a separate window

Figure 1

EMT targeting using NP137. The study by Cassier et al. demonstrated in mice and humans that netrin-1 is up-regulated in endometrial carcinomas. Blocking of netrin-1 by the administration of NP137 led to decreased EMT and reduced tumour progression, validating its clinical potential. Future research should focus on the effects of NP137 in a cardiovascular setting, for example by influencing EndMT. EMT, epithelial-to-mesenchymal transition; EndMT, endothelial-to-mesenchymal transition.

Interestingly, the primary mechanism behind these effects of NP137 was found to be inhibition of EMT, characterized by a decreased expression of mesenchymal genes and increased expression of the epithelial marker EpCAM. Inhibition of EMT occurred not only in mice but also in patients treated with NP137. Furthermore, single-cell RNA sequencing of lung biopsies before and after treatment showed that NP137 significantly reduced the tumour cell compartment. Besides this net decrease in cancer cells, the EMT score was also strongly decreased, demonstrating a more epithelial phenotype. It has already been described that EMT is a major cause of chemotherapy resistance.8 Since NP137 inhibits EMT, Cassier et al. also investigated whether treatment resulted in a more beneficial outcome by comparing carboplatin–pacl*taxel (CarboTaxol), the standard-of-care chemotherapy for the type of malignancy investigated, alone or with addition of NP137. At least in mice, it could be clearly demonstrated that the combined NP137/CarboTaxol treatment was superior to CarboTaxol alone, thus creating the possibility that additive NP137 (on top of other agents) may hold promise for overcoming chemotherapeutic resistance. This notion is currently being further investigated in the Phase 2 GYNET trial (NCT04652076) in patients with endometrial or cervical cancer.

Although great progress has been made over the last years to understand the role of EMT in tumours, so far no pharmacological interventions are available to specifically target EMT in the clinic. Therefore, this study is seminal by proving the clinical potency of NP137. Although the cancer research field is leading the way in efforts to bring the targeting of EMT to the clinic, it is important to extrapolate such observations to other research fields like CVD. For example, intense research efforts have been invested in the identification of compounds that could be used as EndMT inhibitors. Although many of these studies are pre-clinical in nature, several promising approaches have already been identified.9 However, a number of hurdles remain to be overcome, including a lack of robust human data related to EndMT and thus translational proof, and the lack of a standardized definition of EndMT. Furthermore, systemic targeting of EndMT as a therapeutic approach, say for example as a potential approach to stabilize atherosclerotic plaques, would require very careful evaluation due to the likely need for prolonged administration and thus off-target effects. For example, in relation to the current study where EMT is targeted by blocking netrin-1, major side effects may occur in a CVD context as netrin-1 itself has cardioprotective effects after ischaemia–reperfusion injury following a myocardial infarction, which may be lost after netrin-1 blocking approaches.8 Therefore, an approach whereby local delivery could be achieved might be favourable, though still very challenging from a technical perspective.10 Nevertheless, efforts are being made, particularly in pre-clinical studies, to direct nanoparticles to endothelial cells or macrophages, by adding specific peptides to the surface of the particles enabling cell-specific homing. Although such approaches are still in their infancy, it could be speculated that such local delivery methods would also be beneficial in the context of targeting EndMT in atherosclerosis and potentially for other CVDs.

Ultimately, this study from Cassier et al. highlights a long-awaited shift in our outlook towards EMT—being from an interesting biological ‘side observation’ or disease ‘epiphenomenon’, to EMT now being central to disease pathogenesis and a main therapeutic target. While the CVD field is still seemingly years behind that of our oncological colleagues, seen optimistically, this study provides important rationale and motivation to suggest that the targeting of EndMT to ameliorate various CVDs might also someday evolve into an important and unique therapeutic approach.

Contributor Information

Emiel P C van der Vorst, Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany. Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Pauwelsstrasse 17, 52074 Aachen, Germany. Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany. Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Pettenkoferstrasse 9, 80336 Munich, Germany.

Jason C Kovacic, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia. Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. St Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales, Australia.

Funding

E.P.C.v.d.V. is supported by the Interdisciplinary Center for Clinical Research at the RWTH Aachen University. J.C.K. acknowledges the research support from the US National Institutes of Health (R01HL148167), New South Wales Health grant RG194194, the Bourne Foundation, Snow Medical, and Agilent.

Data availability

The manuscript does not contain any new data.

Authors

Therapeutic targeting of cell transition: ready for clinical prime-time? (4)Biography: Dr Emiel van der Vorst obtained his PhD in Cardiovascular Biology and Medicine from the Cardiovascular Research Institute Maastricht (CARIM), The Netherlands in 2015, by studying the effects of high-density lipoproteins on inflammation. For his post-doctoral period, he investigated the role of chemokines and chemokine receptors in atherosclerosis at the Institute for Cardiovascular Prevention (IPEK) in Munich, Germany, resulting in several high-impact publications. Since 2019, he has been working as group leader at the Institute for Molecular Cardiovascular Research (IMCAR) in Aachen, Germany, as well as at CARIM and IPEK. Currently, his research is focusing on elucidating various mechanisms by which lipid metabolism interacts with the immune system in the context of cardiovascular disease, especially atherosclerosis. His work is funded by various prestigious (inter)national personal grants and he is member of the editorial boards of several cardiovascular focused journals. Furthermore, he is a nucleus member of the Scientists of Tomorrow from the European Society of Cardiology.

Therapeutic targeting of cell transition: ready for clinical prime-time? (5)Biography: Professor Jason Kovacic graduated from The University of Melbourne Medical School in 1994 and then undertook cardiology training at St Vincent’s Hospital in Sydney including a PhD in cardiovascular medicine at the Victor Chang Cardiac Research Institute. In 2007, he relocated to the USA, initially to do a post-doc at the National Heart, Lung and Blood Institute (NHLBI) at the National Institutes of Health (NIH) in Bethesda, Maryland. After this post-doc, he moved to The Icahn School of Medicine at Mount Sinai in New York. In early 2020, Jason was appointed as the Institute Director and CEO of the Victor Chang Cardiac Research Institute in Sydney, Australia. He continues to practice as a clinical cardiologist and runs a research programme focused on atherosclerosis, EndMT, fibromuscular dysplasia, and other vascular diseases.

References

1. Evrard SM, Lecce L, Michelis KC, Nomura-Kitabayashi A, Pandey G, Purushothaman K-R, d’Escamard V, Li JR, Hadri L, Fujitani K, Moreno PR, Benard L, Rimmele P, Cohain A, Mecham B, Randolph GJ, Nabel EG, Hajjar R, Fuster V, Boehm M, Kovacic JC. Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat Commun 2016;7:11853. [PMC free article] [PubMed] [Google Scholar]

2. Chen PY, Qin L, Baeyens N, Li G, Afolabi T, Budatha M, Tellides G, Schwartz MA, Simons M. Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest 2015;125(12):4514–4528. [PMC free article] [PubMed] [Google Scholar]

3. Kovacic JC, Dimmeler S, Harvey RP, Finkel T, Aikawa E, Krenning G, Baker AH. Endothelial to mesenchymal transition in cardiovascular disease: JACC state-of-the-art review. J Am Coll Cardiol 2019;73(2):190–209. [PMC free article] [PubMed] [Google Scholar]

4. Hay E. Epithelial-mesenchymal interactions. In: Fleischmajer R, Billingham RE (eds). Proceedings of the 18th Hahnemann Symposium. Philadelphia, PA: Williams and Wilkins; 1968.

5. Trelstad RL, Hay ED, Revel JD. Cell contact during early morphogenesis in the chick embryo. Dev Biol 1967;16(1):78–106. [PubMed] [Google Scholar]

6. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019;20(2):69–84. [PubMed] [Google Scholar]

7. Cassier PA, Navaridas R, Bellina M, Rama N, Ducarouge B, Hernandez-Vargas H, Delord J-P, Lengrand J, Paradisi A, Fattet L, Garin G, Gheit H, Dalban C, Pastushenko I, Neves D, Jelin R, Gadot N, Braissand N, Léon S, Degletagne C, Matias-Guiu X, Devouassoux-Shisheboran M, Mery-Lamarche E, Allard J, Zindy E, Decaestecker C, Salmon I, Perol D, Dolcet X, Ray-Coquard I, Blanpain C, Bernet A, Mehlen P. Netrin-1 blockade inhibits tumour growth and EMT features in endometrial cancer. Nature 2023;620(7973):409–416. [PMC free article] [PubMed] [Google Scholar]

8. Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol 2017;14(10):611–629. [PMC free article] [PubMed] [Google Scholar]

9. Hall I, Kishta F, Xu Y, Baker AH, Kovacic JC. Endothelial to mesenchymal transition: at the axis of cardiovascular health and disease. Cardiovasc Res In Press. [PMC free article] [PubMed] [Google Scholar]

10. Soehnlein O, Libby P. Targeting inflammation in atherosclerosis—from experimental insights to the clinic. Nat Rev Drug Discov 2021;20(8):589–610. [PMC free article] [PubMed] [Google Scholar]

Articles from Cardiovascular Research are provided here courtesy of Oxford University Press

Therapeutic targeting of cell transition: ready for clinical prime-time? (2024)

FAQs

What are the treatment options for mCRC patients? ›

The appropriate adjuvant treatments for mCRC patients are determined by the tumor stage, presence of high-risk pathologic characteristics, microsatellite instability status, patient age, and performance status. Chemotherapy, targeted therapy, and immunotherapy are the main systemic treatments for patients with mCRC.

What is the current treatment for CRC? ›

Treatment usually involves surgical removal of the tumor followed by adjuvant chemotherapy. A clinical trial may also an option. The duration of adjuvant therapy depends on the risk of recurrence (based on characteristics of the cancer that was removed at surgery).

What is the success rate of targeted therapy? ›

While chemotherapy offers around a 30% success rate, targeted therapy is successful in up to 80% of cases.

What are the signs that immunotherapy is working? ›

In general, a positive response to immunotherapy is measured by a shrinking or stable tumor. Although treatment side effects such as inflammation may be a sign that immunotherapy is affecting the immune system in some way, the precise link between immunotherapy side effects and treatment success is unclear.

What is the first line treatment for mCRC? ›

In the first-line setting, pembrolizumab is recommended for patients with mCRC and microsatellite instability-high or deficient mismatch repair tumors; chemotherapy and anti–epidermal growth factor receptor therapy is recommended for microsatellite stable or proficient mismatch repair left-sided treatment-naive RAS ...

What drugs are used to treat mCRPC? ›

Some of the common treatments for mCRPC include chemotherapy (docetaxel), androgen synthesis inhibitors (abiraterone) or androgen signal blockers (enzalutamide, apalutamide). Many patients will ultimately receive second line or third line therapies in the course of their mCRPC treatment.

What is the third line treatment for mCRC? ›

With regard to the third-line treatment of mCRC, either regorafenib 16 or the antimetabolite trifluridine/tipiracil (TAS-102) 17 is recommended in patients irrespective of mutation status.

What is the survival rate of CRC by stage? ›

5-year relative survival rates for colon cancer
SEER stage5-year relative survival rate
Localized91%
Regional72%
Distant13%
All SEER stages combined63%
Jan 29, 2024

What is the mortality rate for CRC? ›

If colon cancer has spread to distant parts of the body, the 5-year relative survival rate is 13%. For rectal cancer, the overall 5-year relative survival rate for people is 68%. If the cancer is diagnosed at a localized stage, the survival rate is 90%.

What if targeted therapy does not work? ›

Cancer cells can eventually stop responding to a targeted therapy drug even if it works at first. If this happens, another targeted therapy drug or another treatment may be offered. Less commonly, a targeted therapy drug may cause serious side effects and the treatment plan will need to be adjusted.

Does targeted therapy always work? ›

A targeted treatment will not work if the tumor does not have the specific genetic change or protein the drug targets. Even if you have the specific genetic change or protein the drug targets, it's possible the tumor will not respond to the drug. The response to the treatment may not last over time.

What are the disadvantages of targeted therapy? ›

Some of the common side effects of targeted therapy may include:
  • allergic reactions.
  • anemia and low blood counts.
  • birth defects.
  • fatigue.
  • hair loss.
  • heart damage.
  • joint or muscle pain.
  • nausea, diarrhea, vomiting and appetite changes, and weight loss.

How sick does immunotherapy make you? ›

Some of the most common side effects associated with immunotherapy treatment may include but are not limited to: chills, constipation, coughing, decreased appetite, diarrhea, fatigue, fever and flu-like symptoms, headache, infusion-related reaction or injection site pain, itching, localized rashes and/or blisters, ...

Why do I feel worse after immunotherapy? ›

Although immunotherapy is designed to help your immune system attack cancer cells, immune cells may mistakenly attack healthy tissue. This is called an immune-related adverse effect, or irAE. As a result, immunotherapy can cause inflammation in healthy tissue that you may experience as one or more side effects.

Is there any other therapy for mcrpc? ›

Second-line hormone therapies like abiraterone and enzalutamide. These target male hormones in different ways than first-line ADT. These oral drugs can extend life. Radioactive material like Xofigo (radium-223 dichloride).

What is the second-line treatment for mCRC? ›

The second-line regimen choice depends on the systemic therapies given in first line. FOLFOX or XELOX are recommended for those who failed in irinotecan-based CT in the first line, and oxaliplatin-based CT are suggested for those who have received irinotecan-based CT regimen.

What is the standard of care for mCRC? ›

The current standard of care for first-line treatment is combination cytotoxic chemotherapy using the fluoropyrimidine backbone (5-FU or capecitabine) with either oxaliplatin (FOLFOX or XELOX) or irinotecan (FOLFIRI or XELIRI) in combination with the anti-VEGF agent bevacizumab or anti-EGFR agents (cetuximab or ...

References

Top Articles
Latest Posts
Article information

Author: Jamar Nader

Last Updated:

Views: 6061

Rating: 4.4 / 5 (55 voted)

Reviews: 94% of readers found this page helpful

Author information

Name: Jamar Nader

Birthday: 1995-02-28

Address: Apt. 536 6162 Reichel Greens, Port Zackaryside, CT 22682-9804

Phone: +9958384818317

Job: IT Representative

Hobby: Scrapbooking, Hiking, Hunting, Kite flying, Blacksmithing, Video gaming, Foraging

Introduction: My name is Jamar Nader, I am a fine, shiny, colorful, bright, nice, perfect, curious person who loves writing and wants to share my knowledge and understanding with you.