Flyer

Archives in Cancer Research

  • ISSN: 2254-6081
  • Journal h-index: 14
  • Journal CiteScore: 3.77
  • Journal Impact Factor: 4.09
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Awards Nomination 20+ Million Readerbase
Indexed In
  • China National Knowledge Infrastructure (CNKI)
  • CiteFactor
  • OCLC- WorldCat
  • Publons
  • Geneva Foundation for Medical Education and Research
  • Euro Pub
  • Google Scholar
  • J-Gate
  • Secret Search Engine Labs
  • International Committee of Medical Journal Editors (ICMJE)
  • Zenodo
Share This Page

Mini Review - (2022) Volume 10, Issue 9

New methods for studying cancer metastases using metabolomics

David Cruzado*
 
Department of Medical Oncology, Dana- Farber Cancer Institute, Boston, MA, USA
 
*Correspondence: David Cruzado, Department of Medical Oncology, Dana- Farber Cancer Institute, Boston, MA, USA, Email:

Received: 01-Sep-2022, Manuscript No. ipacr-22-13017; Editor assigned: 05-Sep-2022, Pre QC No. ipacr-22-13017; Reviewed: 12-Sep-2022, QC No. ipacr-22-13017; Revised: 19-Sep-2022, Manuscript No. ipacr-22-13017 ; Published: 29-Sep-2022, DOI: 10.36648/2254-6081-10.9-146

Abstract

Metastasis is accountable for ninetieth of deaths in patients with cancer. Understanding the role of metabolism throughout metastasis has been restricted by the event of strong and sensitive technologies that capture metabolic processes in metastasizing cancer cells. We have a tendency to discuss the present technologies out there to check metabolism in primary and pathologic process cancer cells and metabolic interactions between cancer cells and also the neoplasm microenvironment (TME) at totally different stages of the pathologic process cascade. We have a tendency to establish benefits and downsides of every methodology and discuss however these tools and technologies can more improve our understanding of metastasis. Studies investigation the advanced metabolic rewiring of various cells victimization progressive metabolomic technologies have the potential to reveal novel biological processes and therapeutic interventions for human cancers.

Keywords

Cancer metastases; Tumour micro-environment; Drug delivery; Stimulus-response; Metabolomics

Introduction

Metabolism is central to all or any cellular functions, as cellular metabolism ultimately drives the generation and usage of adenosine triphosphate needed for cellular activity. Changes at the metabolic level will result in many diseases, together with cancer [1,2]. Cancer cells will activate and suppress totally different metabolic pathways. Moreover, metabolic changes known in cancer cells compared with non-cancer cells provide potential metabolic vulnerabilities which will be therapeutically modulated to decrease cancer progression. Over the past few years, there has been exceptional progress in understanding the metabolic regulation of cancer cells, particularly within the context of primary neoplasm metabolism. However, a comprehensive understanding of the metabolic regulation of cancer cells throughout metastasis (see Glossary) remains AN open space of active analysis.

Metastasis could be a advanced method that involves uncontrolled proliferation of cancer cells at a primary web site, native invasion into encompassing tissue, trauma into the blood or lymphatic’s, transportation through circulation extravasation into distant tissues, and colonisation of secondary organs [3,4]. Metastasis through this {metastatic pathologic method pathological process} cascade could be a extremely inefficient process as most cancer cells don't survive, however some bear metabolic variations to optimize survival in these harsh environments. Metastasis is characterised by the flexibility of cancer cells to maneuver from their primary web site and spread through the blood or humour to make a neoplasm in distant organs [5]. the event of metastasis needs cancer cells to initial invade into the tumourassociated stroma And bear an animal tissue-to-mesenchymal transition (EMT) inflicting epithelial cells to lose their polarity and gain the flexibility to invade, resist stress, and spread with a mesenchymal-like makeup many metabolites together with 2-hydroxyglutarate are shown to modulate EMT through the modulation of transcription factors when no-hit invasion, cancer cells initiate the method of trauma whereby cancer cells elongate, generating protrusions that enable cells to pass between the epithelial tissue cells of the blood and bodily fluid vessels and migrate into the circulation [6]. Rising proof indicates that some metabolites area unit proangiogenic and prolymphangiogenic, so increasing vessel density within the primary neoplasm, supply it with gas and nutrients, maintaining the neoplasm metabolism, and promoting metastasis. Once within the circulation, current neoplasm cells (CTCs) area unit exposed to setting stress together with aerophilous stress and ferroptosis and area unit forced to metabolically adapt to survive in such a harsh environment.

Sensitive and sturdy metabolomic technologies area unit vital for detection the metabolic rewiring of the few cancer cells out there for study at numerous steps of the pathologic process cascade from: heterogeneous primary tumors current neoplasm cells (CTCs), and distant pathologic process organ sites thanks to enhancements in instrumentation, it's currently doable to investigate the metabolomic profile of little numbers of cancer cells during this review, we have a tendency to summarize the newest metabolomic technologies out there to investigate neoplastic cell metabolism, with a stress on metastasizing cancer cells in diagnosis models and in patients. We have a tendency to discuss the benefits and downsides of every methodology and challenges to be overcome. Elucidating the metabolic vulnerabilities of cancer throughout metastasis is key for the event of latest therapies to dam metastasis formation. Quantitative metabolomics has long been a robust analytic tool for evaluating metabolites in biological samples. Just like several ‘-omics’ sciences, metabolomic technologies area unit perpetually evolving, driving new developments in analytical techniques, models, software, and procedure ways to enhance sensitivity and specificity. Techniques developed decades past like proton magnetic resonance spectrographic analysis, gas chromatography–mass spectroscopy (GC-MS), and liquid chromatography–MS (LC-MS) have verified to be vital tools for the detection and quantification of metabolites in cancer metastasis [7,8]. However, these techniques need a comparatively sizable amount of cancer cells (>10 000 cells) and can't distinguish the metabolic heterogeneousness on a singlecell level. Characterizing the metabolic heterogeneousness at intervals a neoplasm is very important as variations in subsets of willcer cells at intervals a neoplasm can influence the extent of economical metastasis and potential response to therapies. The presence or absence of specific metabolic qualities in subsets of cancer cells is also helpful for predicting that cancer cells area unit doubtless to expeditiously spread, and so might function biomarkers for diagnostic prognosis

Discussion

Single-cell analysis is a very promising technology that has insight into cellular heterogeneousness and dynamics in individual cells and has been used to live transcriptome, proteome, and metabolomic abundance In primary and pathologic process tumors, single-cell metabolomics (SCM) is wont to reveal metabolic heterogeneousness at intervals a neoplastic cell population. Moreover, SCM is useful for uncovering distinct metabolic profiles of individual cell varieties at intervals a neoplasm (i.e., immune cells and stromal cells) and has been applied to find variations needed for metastasis or for medical care resistance. SCM has been employed in pathologic process malignant melanoma and first and pathologic process neoplasms of head and neck cancers to demonstrate that neoplasm and non-tumour cells at intervals [9]. the TME have totally different metabolic activity that weren't detected in bulk tumour tissues. SCM, still as low-cell range metabolomics, is especially helpful within the analysis of CTCs in blood thanks to high intrapatient heterogeneousness and pertinence as a liquid diagnostic assay biomarker for metastasis for instance; current malignant melanoma cells were shown to own down regulated purine synthesis compared with primary neoplasm cells.

However, metabolic analysis of CTCs continues to be difficult. Isolation of CTCs is usually performed during a flow cytometer. However, the isolation method will cause metabolic stress to the individual cells thanks to shear forces and pressure. Therefore, direct sorting into correct ending solutions (e.g., methanol) and short time periods area unit counselled to avoid metabolic disturbances [10]. An alternate for enrichment of CTCs is that the ParsortixTM Cell Separation container System, a micro fluid platform that captures single CTCs supported size, that's a gentler methodology compared with flow cytometer. However, this microfluid system has lower flow-through compared with flow cytometer, and there are limitations to the numbers of CTCs that may be collected in one container. In summary, SCM could be a speedily progressing field with recent important advances in sturdy sampling, ionization ways, and substance detection. Continuing development of sample preparation protocols to extend the metabolic integrity of analysed cells can provide further enhancements to SCM analyses.

Changes in matrix chemistry and advanced instrumentation have allowed MALDI-MSI to be applied in cancer studies to quantify metabolites, discover biomarkers for medicine and prognostics, and perceive drug bioavailability at intervals tumors. Recently, demonstrating that pathologic process breast cancers have higher levels of N-glycan’s compared with primary tumors. That there area unit fewer phospholipids in stroma and non-cancer epithelial tissue compared to prostatic adenocarcinoma cells

Isotope tracing is characterised by the pursuit of nutrients, tagged with stable or hot isotopes, that area unit more analysed via MS and/or proton magnetic resonance. This methodology identifies wherever atoms from the infused substrate (tracer) area unit metabolized to (tracee). Therefore, atom tracing provides elaborated data on contributions to specific pathways, that is, a substrate being metabolized through the simple sugar phosphate pathway, etc. To quantitate and interpret atom tracing studies, there area unit many conditions that area unit required the infusion of atom tracer should be at atom steady state.

Conclusion

The field of cancer metabolomics analysis has greatly evolved with the event of the new technologies and tools highlighted during this review. These technologies have helped to spot metabolic alterations in primary tumors and distant pathologic process sites that are applied within the clinic for cancer medicine and medical care. However, the foremost recent advances in these technologies have allowed the analysis of metastasizing cancer cells. These techniques have the potential to uncover why just some cancer cells survive within the blood throughout metastasis, however their metabolism changes, and why they spread to specific secondary organs. Still, enhancements in substance detection sensitivity, characterization of ‘malignant’ metabolic profiles of metastasizing cancer cells for patient stratification, prognosis, and medical care, stay to be elucidated (see Outstanding questions). Over consecutive decade, uncovering and targeting the metabolic pathways in metastasizing cancer cells can still reveal new therapeutic targets with potential for reducing pathologic process unfold to enhance patient prognosis and survival.

Acknowledgement

I would like to thank my professor for his support and encouragement.

Conflict of Interest

The authors declare that there is no conflict of interest.

Keywords

Cancer metastases; Tumour micro-environment; Drug delivery; Stimulus-response; Metabolomics

Introduction

Metabolism is central to all or any cellular functions, as cellular metabolism ultimately drives the generation and usage of adenosine triphosphate needed for cellular activity. Changes at the metabolic level will result in many diseases, together with cancer [1,2]. Cancer cells will activate and suppress totally different metabolic pathways. Moreover, metabolic changes known in cancer cells compared with non-cancer cells provide potential metabolic vulnerabilities which will be therapeutically modulated to decrease cancer progression. Over the past few years, there has been exceptional progress in understanding the metabolic regulation of cancer cells, particularly within the context of primary neoplasm metabolism. However, a comprehensive understanding of the metabolic regulation of cancer cells throughout metastasis (see Glossary) remains AN open space of active analysis.

Metastasis could be a advanced method that involves uncontrolled proliferation of cancer cells at a primary web site, native invasion into encompassing tissue, trauma into the blood or lymphatic’s, transportation through circulation extravasation into distant tissues, and colonisation of secondary organs [3,4]. Metastasis through this {metastatic pathologic method pathological process} cascade could be a extremely inefficient process as most cancer cells don't survive, however some bear metabolic variations to optimize survival in these harsh environments. Metastasis is characterised by the flexibility of cancer cells to maneuver from their primary web site and spread through the blood or humour to make a neoplasm in distant organs [5]. the event of metastasis needs cancer cells to initial invade into the tumourassociated stroma And bear an animal tissue-to-mesenchymal transition (EMT) inflicting epithelial cells to lose their polarity and gain the flexibility to invade, resist stress, and spread with a mesenchymal-like makeup many metabolites together with 2-hydroxyglutarate are shown to modulate EMT through the modulation of transcription factors when no-hit invasion, cancer cells initiate the method of trauma whereby cancer cells elongate, generating protrusions that enable cells to pass between the epithelial tissue cells of the blood and bodily fluid vessels and migrate into the circulation [6]. Rising proof indicates that some metabolites area unit proangiogenic and prolymphangiogenic, so increasing vessel density within the primary neoplasm, supply it with gas and nutrients, maintaining the neoplasm metabolism, and promoting metastasis. Once within the circulation, current neoplasm cells (CTCs) area unit exposed to setting stress together with aerophilous stress and ferroptosis and area unit forced to metabolically adapt to survive in such a harsh environment.

Sensitive and sturdy metabolomic technologies area unit vital for detection the metabolic rewiring of the few cancer cells out there for study at numerous steps of the pathologic process cascade from: heterogeneous primary tumors current neoplasm cells (CTCs), and distant pathologic process organ sites thanks to enhancements in instrumentation, it's currently doable to investigate the metabolomic profile of little numbers of cancer cells during this review, we have a tendency to summarize the newest metabolomic technologies out there to investigate neoplastic cell metabolism, with a stress on metastasizing cancer cells in diagnosis models and in patients. We have a tendency to discuss the benefits and downsides of every methodology and challenges to be overcome. Elucidating the metabolic vulnerabilities of cancer throughout metastasis is key for the event of latest therapies to dam metastasis formation. Quantitative metabolomics has long been a robust analytic tool for evaluating metabolites in biological samples. Just like several ‘-omics’ sciences, metabolomic technologies area unit perpetually evolving, driving new developments in analytical techniques, models, software, and procedure ways to enhance sensitivity and specificity. Techniques developed decades past like proton magnetic resonance spectrographic analysis, gas chromatography–mass spectroscopy (GC-MS), and liquid chromatography–MS (LC-MS) have verified to be vital tools for the detection and quantification of metabolites in cancer metastasis [7,8]. However, these techniques need a comparatively sizable amount of cancer cells (>10 000 cells) and can't distinguish the metabolic heterogeneousness on a singlecell level. Characterizing the metabolic heterogeneousness at intervals a neoplasm is very important as variations in subsets of willcer cells at intervals a neoplasm can influence the extent of economical metastasis and potential response to therapies. The presence or absence of specific metabolic qualities in subsets of cancer cells is also helpful for predicting that cancer cells area unit doubtless to expeditiously spread, and so might function biomarkers for diagnostic prognosis

Discussion

Single-cell analysis is a very promising technology that has insight into cellular heterogeneousness and dynamics in individual cells and has been used to live transcriptome, proteome, and metabolomic abundance In primary and pathologic process tumors, single-cell metabolomics (SCM) is wont to reveal metabolic heterogeneousness at intervals a neoplastic cell population. Moreover, SCM is useful for uncovering distinct metabolic profiles of individual cell varieties at intervals a neoplasm (i.e., immune cells and stromal cells) and has been applied to find variations needed for metastasis or for medical care resistance. SCM has been employed in pathologic process malignant melanoma and first and pathologic process neoplasms of head and neck cancers to demonstrate that neoplasm and non-tumour cells at intervals [9]. the TME have totally different metabolic activity that weren't detected in bulk tumour tissues. SCM, still as low-cell range metabolomics, is especially helpful within the analysis of CTCs in blood thanks to high intrapatient heterogeneousness and pertinence as a liquid diagnostic assay biomarker for metastasis for instance; current malignant melanoma cells were shown to own down regulated purine synthesis compared with primary neoplasm cells.

However, metabolic analysis of CTCs continues to be difficult. Isolation of CTCs is usually performed during a flow cytometer. However, the isolation method will cause metabolic stress to the individual cells thanks to shear forces and pressure. Therefore, direct sorting into correct ending solutions (e.g., methanol) and short time periods area unit counselled to avoid metabolic disturbances [10]. An alternate for enrichment of CTCs is that the ParsortixTM Cell Separation container System, a micro fluid platform that captures single CTCs supported size, that's a gentler methodology compared with flow cytometer. However, this microfluid system has lower flow-through compared with flow cytometer, and there are limitations to the numbers of CTCs that may be collected in one container. In summary, SCM could be a speedily progressing field with recent important advances in sturdy sampling, ionization ways, and substance detection. Continuing development of sample preparation protocols to extend the metabolic integrity of analysed cells can provide further enhancements to SCM analyses.

Changes in matrix chemistry and advanced instrumentation have allowed MALDI-MSI to be applied in cancer studies to quantify metabolites, discover biomarkers for medicine and prognostics, and perceive drug bioavailability at intervals tumors. Recently, demonstrating that pathologic process breast cancers have higher levels of N-glycan’s compared with primary tumors. That there area unit fewer phospholipids in stroma and non-cancer epithelial tissue compared to prostatic adenocarcinoma cells

Isotope tracing is characterised by the pursuit of nutrients, tagged with stable or hot isotopes, that area unit more analysed via MS and/or proton magnetic resonance. This methodology identifies wherever atoms from the infused substrate (tracer) area unit metabolized to (tracee). Therefore, atom tracing provides elaborated data on contributions to specific pathways, that is, a substrate being metabolized through the simple sugar phosphate pathway, etc. To quantitate and interpret atom tracing studies, there area unit many conditions that area unit required the infusion of atom tracer should be at atom steady state.

Conclusion

The field of cancer metabolomics analysis has greatly evolved with the event of the new technologies and tools highlighted during this review. These technologies have helped to spot metabolic alterations in primary tumors and distant pathologic process sites that are applied within the clinic for cancer medicine and medical care. However, the foremost recent advances in these technologies have allowed the analysis of metastasizing cancer cells. These techniques have the potential to uncover why just some cancer cells survive within the blood throughout metastasis, however their metabolism changes, and why they spread to specific secondary organs. Still, enhancements in substance detection sensitivity, characterization of ‘malignant’ metabolic profiles of metastasizing cancer cells for patient stratification, prognosis, and medical care, stay to be elucidated (see Outstanding questions). Over consecutive decade, uncovering and targeting the metabolic pathways in metastasizing cancer cells can still reveal new therapeutic targets with potential for reducing pathologic process unfold to enhance patient prognosis and survival.

Acknowledgement

I would like to thank my professor for his support and encouragement.

Conflict of Interest

The authors declare that there is no conflict of interest.

REFERENCES

  1. Inmaculada M, Navdeep S C (2021) Cancer metabolism: looking forward. Nat Rev Cancer21: 669-680.
  2. Indexed at, Google Scholar, Crossref

  3. Tom M J E, Mazène H, Sander J T, Ron M A H, Stefan S (2019) Deciphering metabolic heterogeneity by single-cell analysis. Anal Chem 91: 13314-13323.
  4. Indexed at, Google Scholar, Crossref

  5. Arthur W L, Diwakar R P, Robert A W (2017) Emerging Biological Principles of Metastasis. Cell168: 670-691.
  6. Indexed at, Google Scholar, Crossref

  7. Ralph J D, Navdeep S C (2020) We need to talk about the Warburg effect. Nat Metab 2: 127-129.
  8. Indexed at, Google Scholar, Crossref

  9. Warburg O, Wind F, Negelein E (1927) The metabolism of tumors in the body. J Gen Physiol8: 519-530.
  10. Indexed at, Google Scholar, Crossref

  11. Jennifer G G, Piskounova E , Sean J M (2016) Cancer, Oxidative Stress, and Metastasis. Cold Spring Harb Symp Quant Biol81: 163-175.
  12. Indexed at, Google Scholar, Crossref

  13. Patrick M, Alain P (2006) Metastasis: a question of life or death. Nat Rev Cancer6: 449-458.
  14. Indexed at, Google Scholar, Crossref

  15. Alpaslan T, Brandon F, Vijayashree R, Jessalyn M U, Shen B et al. (2020) Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature577: 115-120.
  16. Indexed at, Google Scholar, Crossref

  17. Gabriele B, Sarah M F (2021) The metabolism of cancer cells during metastasis. Nat Rev Cancer21: 162-180.
  18. Indexed at, Google Scholar, Crossref

  19. Karuna G, Joan M (2021) Targeting metastatic cancer. Nat Med27: 34-44.
  20. Indexed at, Google Scholar, Crossref

Citation: Cruzado D (2022) New methods for studying cancer metastases using metabolomics. Archives Can Res, Vol.10 No. Issue 9:146.