Next Article in Journal
Early Determinants of Adverse Motor Outcomes in Preschool Children with a Critical Congenital Heart Defect
Next Article in Special Issue
Pathways of Angiogenic and Inflammatory Cytokines in Multiple Myeloma: Role in Plasma Cell Clonal Expansion and Drug Resistance
Previous Article in Journal
Measures of Corticalization
Previous Article in Special Issue
A Proliferation-Inducing Ligand and B-Cell Activating Factor Are Upregulated in Patients with Essential Thrombocythemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

The Urgent Need for Precision Medicine in Cancer and Its Microenvironment: The Paradigmatic Case of Multiple Myeloma

by
Antonio Giovanni Solimando
1,*,
Markus Krebs
2,3,
Max Bittrich
4 and
Hermann Einsele
4
1
Guido Baccelli Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology, School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy
2
Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
3
Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080 Würzburg, Germany
4
Department of Internal Medicine II, University Hospital Würzburg, Josef-Schneider Straße 2, 97080 Würzburg, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(18), 5461; https://doi.org/10.3390/jcm11185461
Submission received: 12 September 2022 / Accepted: 15 September 2022 / Published: 16 September 2022
(This article belongs to the Special Issue Immune Microenvironment in Solid and Hematological Malignancies)
Precision medicine is particularly relevant for cancer and microenvironment deconvolution for therapeutic purposes in hematological and non-hematological malignancies [1,2,3,4]. Multiple myeloma (MM) is no exception, representing a paradigmatic condition. Indeed, although the advent of next-generation sequencing uncovered the genomic makeup, MM is a disease characterized by high genetic complexity [5,6]. Primary events, including IgH translocations and hyperdiploidy, and secondary events such as CNV, SNP structural variations, and epigenetic changes also occur [7,8,9]. Intra-patient/intratumoral and interpatient heterogeneity, given the subclonal population’s extreme dynamics, starts from the beginning and the asymptomatic condition through all the stages of the disease [10,11,12]. On top of this, treatment selection further boosts the disease complexity, while prompting novel therapeutic strategies [13,14]. Treatment options are also heterogeneous [15]. The large number of therapeutic alternatives currently available for MM patients gives rise to a wide range of possible clinical settings that cannot be easily covered by a single algorithm [16].
Multimodality spans clinical, molecular, and imaging levels. Several factors such as demographics, biochemical properties, staging, tumor burden (BMPC), minimal measurable disease, transplant status, treatments, and outcomes should be combined with cytogenetics, RNAseq, microarray, and novel tools (such as scRNASeq, WES, WGS, targeted sequencing, methylation, and proteomics). Imaging techniques, such as PET-CT scans and MRIs are also relevant. This landscape represents a typical framework for a machine learning approach [5]. The main issue of the existing valuable datasets in the literature is that they are very sparse regarding the specific modalities [17]. The attempts made to define simple features with high predictive power have been validated in the investigations regarding the progression from MGUS/SMM to MM or the survival risk [18,19,20]. Despite being effective, these simple, unimodal models lack effectiveness in some patient subgroups, namely the ultra-high-risk patients [21]. Thus, crowdsourced efforts have been made to develop machine learning models of rapid clinical progression by 18 months in newly diagnosed MM patients using a combination of DNA-, RNA-, age, ISS, and other demographic, clinical, or cytogenetic-based features [22]. Mason et al. benchmarked their machine learning approach against previously published models. In this DREAM challenge study, the data-driven risk predictive models leveraged high-dimensional, multimodal, and multiexperimental data, and the analysis of top-performing methods identified the high expression of PHF19 as the gene with the strongest association with MM progression [22]. Although an attempt to address patient subgroup stratification has also been made [17], validation is largely needed in real life. Collectively, the challenges for precision medicine in MM are constituted by the heterogeneity of the disease, also considering the treatments available in clinical practice. Mostly, the existing datasets are “small”, soiled, and sparse, and multimodal resources are still lacking. Therefore, multimodal and federated approaches to breaking data silos are mainly needed [23].
An integrative modeling approach might represent a possible integration of the data and prior biomedical knowledge to overcome data limitations [24,25]. This approach would imply a shift from a gene-centric to a network approach. Multiple myeloma represents a landscape in which more precise identification of high-risk disease relapse has been possible based on these novel approaches [26]. Based on the transcriptome, Wall et al. developed a complex pipeline eventually able to associate a more efficient signature with each subgroup, in terms of network activity rather than gene activity, with the advantage of being more interpretable [26].
Similar approaches have also been applied to more complicated projects, namely a personalized assessment of the risk of progression from MGUS/SMM to MM [27]. These studies highlight that even though simple predictors (such as age) perform well, the specific landscape of the genomic makeup characterizes stable and progressive subgroups [27]. One major challenge is related to the lack of access to a high volume of content data in the pre-malignant phases.
There is an urgent need for precision medicine in cancer and multiple myeloma [28]. The sparsity of datasets in the literature represents significant constraints on crucial modalities on single datasets compared with the disease’s level of heterogeneity. Machine learning models have already been proven valuable as complementary tools to well-established clinical and bioinformatic approaches. By leveraging multimodalities and prior biomedical knowledge, artificial intelligence can provide a more efficient risk score and suggest new biomarkers and subgroup–treatment associations [22,29,30].
Overall, we are confident that the issues raised in this editorial might help to interpret the findings of the current clinical and pre-clinical practice. The authors plan to generate questions about the optimal treatment approach for cancer therapy and its microenvironment, considering the novel tools presented.

Acknowledgments

We thank Mary Victoria Pragnell, in the School of Medicine and Surgery at the University of Bari for professional linguistic revision and professional editing.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Bolkun, L.; Tynecka, M.; Wasiluk, T.; Piszcz, J.; Starosz, A.; Grubczak, K.; Moniuszko, M.; Eljaszewicz, A. A Proliferation-Inducing Ligand and B-Cell Activating Factor Are Upregulated in Patients with Essential Thrombocythemia. JCM 2022, 11, 4663. [Google Scholar] [CrossRef]
  2. Rasche, L.; Hudecek, M.; Einsele, H. What Is the Future of Immunotherapy in Multiple Myeloma? Blood 2020, 136, 2491–2497. [Google Scholar] [CrossRef]
  3. Clara, J.A.; Monge, C.; Yang, Y.; Takebe, N. Targeting Signalling Pathways and the Immune Microenvironment of Cancer Stem Cells—A Clinical Update. Nat. Rev. Clin. Oncol. 2020, 17, 204–232. [Google Scholar] [CrossRef]
  4. Argentiero, A.; Solimando, A.G.; Krebs, M.; Leone, P.; Susca, N.; Brunetti, O.; Racanelli, V.; Vacca, A.; Silvestris, N. Anti-Angiogenesis and Immunotherapy: Novel Paradigms to Envision Tailored Approaches in Renal Cell-Carcinoma. J. Clin. Med. 2020, 9, 594. [Google Scholar] [CrossRef] [PubMed]
  5. Dutta, A.K.; Alberge, J.-B.; Sklavenitis-Pistofidis, R.; Lightbody, E.D.; Getz, G.; Ghobrial, I.M. Single-Cell Profiling of Tumour Evolution in Multiple Myeloma—Opportunities for Precision Medicine. Nat. Rev. Clin. Oncol. 2022, 19, 223–236. [Google Scholar] [CrossRef] [PubMed]
  6. Desantis, V.; Savino, F.D.; Scaringella, A.; Potenza, M.A.; Nacci, C.; Frassanito, M.A.; Vacca, A.; Montagnani, M. The Leading Role of the Immune Microenvironment in Multiple Myeloma: A New Target with a Great Prognostic and Clinical Value. JCM 2022, 11, 2513. [Google Scholar] [CrossRef] [PubMed]
  7. Morgan, G.J.; Walker, B.A.; Davies, F.E. The Genetic Architecture of Multiple Myeloma. Nat. Rev. Cancer 2012, 12, 335–348. [Google Scholar] [CrossRef]
  8. Da Via’, M.C.; Solimando, A.G.; Garitano-Trojaola, A.; Barrio, S.; Rodhes, N.; Strifler, S.; Teufel, E.; Lapa, C.; Einsele, H.; Beilhack, A.; et al. CIC-Mutation as a Potential Molecular Mechanism of Acquired Resistance to Combined BRAF/MEK Inhibition in CNS Multiple Myeloma. Blood 2018, 132, 3181. [Google Scholar] [CrossRef]
  9. Sallustio, F.; Curci, C.; Solimando, A.G.; Leone, P.; Pontrelli, P.; Gesualdo, L.; Vacca, A.; Racanelli, V.; Gallone, A. Identification and Monitoring of Copy Number Variants (CNV) in Monoclonal Gammopathy. Cancer Biol. Ther. 2021, 22, 404–412. [Google Scholar] [CrossRef]
  10. Schürch, C.M.; Rasche, L.; Frauenfeld, L.; Weinhold, N.; Fend, F. A Review on Tumor Heterogeneity and Evolution in Multiple Myeloma: Pathological, Radiological, Molecular Genetics, and Clinical Integration. Virchows Arch. 2020, 476, 337–351. [Google Scholar] [CrossRef]
  11. Solimando, A.G.; Da Vià, M.C.; Bolli, N.; Steinbrunn, T. The Route of the Malignant Plasma Cell in Its Survival Niche: Exploring “Multiple Myelomas”. Cancers 2022, 14, 3271. [Google Scholar] [CrossRef] [PubMed]
  12. Rasche, L.; Schinke, C.; Maura, F.; Bauer, M.A.; Ashby, C.; Deshpande, S.; Poos, A.M.; Zangari, M.; Thanendrarajan, S.; Davies, F.E.; et al. The Spatio-Temporal Evolution of Multiple Myeloma from Baseline to Relapse-Refractory States. Nat. Commun. 2022, 13, 4517. [Google Scholar] [CrossRef] [PubMed]
  13. Mikkilineni, L.; Kochenderfer, J.N. CAR T Cell Therapies for Patients with Multiple Myeloma. Nat. Rev. Clin. Oncol. 2021, 18, 71–84. [Google Scholar] [CrossRef] [PubMed]
  14. Desantis, V.; Saltarella, I.; Lamanuzzi, A.; Melaccio, A.; Solimando, A.G.; Mariggiò, M.A.; Racanelli, V.; Paradiso, A.; Vacca, A.; Frassanito, M.A. MicroRNAs-Based Nano-Strategies as New Therapeutic Approach in Multiple Myeloma to Overcome Disease Progression and Drug Resistance. Int. J. Mol. Sci. 2020, 21, 3084. [Google Scholar] [CrossRef]
  15. Gulla, A.; Anderson, K.C. Multiple Myeloma: The (r)Evolution of Current Therapy and a Glance into Future. Haematologica 2020, 105, 2358–2367. [Google Scholar] [CrossRef]
  16. Hernández-Rivas, J.-Á.; Ríos-Tamayo, R.; Encinas, C.; Alonso, R.; Lahuerta, J.-J. The Changing Landscape of Relapsed and/or Refractory Multiple Myeloma (MM): Fundamentals and Controversies. Biomark. Res. 2022, 10, 1. [Google Scholar] [CrossRef]
  17. Bhalla, S.; Melnekoff, D.T.; Aleman, A.; Leshchenko, V.; Restrepo, P.; Keats, J.; Onel, K.; Sawyer, J.R.; Madduri, D.; Richter, J.; et al. Patient Similarity Network of Newly Diagnosed Multiple Myeloma Identifies Patient Subgroups with Distinct Genetic Features and Clinical Implications. Sci. Adv. 2021, 7, eabg9551. [Google Scholar] [CrossRef]
  18. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.-V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International Myeloma Working Group Updated Criteria for the Diagnosis of Multiple Myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef]
  19. Ho, M.; Patel, A.; Goh, C.Y.; Moscvin, M.; Zhang, L.; Bianchi, G. Changing Paradigms in Diagnosis and Treatment of Monoclonal Gammopathy of Undetermined Significance (MGUS) and Smoldering Multiple Myeloma (SMM). Leukemia 2020, 34, 3111–3125. [Google Scholar] [CrossRef]
  20. Perrot, A.; Corre, J.; Avet-Loiseau, H. Risk Stratification and Targets in Multiple Myeloma: From Genomics to the Bedside. Am. Soc. Clin. Oncol. Educ. Book 2018, 38, 675–680. [Google Scholar] [CrossRef]
  21. Solimando, A.G.; Da Vià, M.C.; Cicco, S.; Leone, P.; Di Lernia, G.; Giannico, D.; Desantis, V.; Frassanito, M.A.; Morizio, A.; Delgado Tascon, J.; et al. High-Risk Multiple Myeloma: Integrated Clinical and Omics Approach Dissects the Neoplastic Clone and the Tumor Microenvironment. J. Clin. Med. 2019, 8, 997. [Google Scholar] [CrossRef] [PubMed]
  22. Mason, M.J.; Schinke, C.; Eng, C.L.P.; Towfic, F.; Gruber, F.; Dervan, A.; White, B.S.; Pratapa, A.; Guan, Y.; Chen, H.; et al. Multiple Myeloma DREAM Challenge Reveals Epigenetic Regulator PHF19 as Marker of Aggressive Disease. Leukemia 2020, 34, 1866–1874. [Google Scholar] [CrossRef] [PubMed]
  23. Du Terrail, J.O.; Léopold, A.; Joly, C.; Beguier, C.; Andreux, M.; Maussion, C.; Schmauch, B.; Tramel, E.W.; Bendjebbar, E.; Zaslavskiy, M.; et al. Collaborative Federated Learning behind Hospitals’ Firewalls for Predicting Histological Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. J. Clin. Oncol. 2021, 40, 590. [Google Scholar] [CrossRef]
  24. Hofree, M.; Shen, J.P.; Carter, H.; Gross, A.; Ideker, T. Network-Based Stratification of Tumor Mutations. Nat. Methods 2013, 10, 1108–1115. [Google Scholar] [CrossRef]
  25. Geng, Y.; Chen, J.; Jimenez-Ruiz, E.; Chen, H. Human-Centric Transfer Learning Explanation via Knowledge Graph [Extended Abstract]. arxiv 2019, arXiv:1901.08547. [Google Scholar] [CrossRef]
  26. Wall, M.A.; Turkarslan, S.; Wu, W.-J.; Danziger, S.A.; Reiss, D.J.; Mason, M.J.; Dervan, A.P.; Trotter, M.W.B.; Bassett, D.; Hershberg, R.M.; et al. Genetic Program Activity Delineates Risk, Relapse, and Therapy Responsiveness in Multiple Myeloma. NPJ Precis. Oncol. 2021, 5, 60. [Google Scholar] [CrossRef]
  27. Oben, B.; Froyen, G.; Maclachlan, K.H.; Leongamornlert, D.; Abascal, F.; Zheng-Lin, B.; Yellapantula, V.; Derkach, A.; Geerdens, E.; Diamond, B.T.; et al. Whole-Genome Sequencing Reveals Progressive versus Stable Myeloma Precursor Conditions as Two Distinct Entities. Nat. Commun. 2021, 12, 1861. [Google Scholar] [CrossRef]
  28. Saltarella, I.; Desantis, V.; Melaccio, A.; Solimando, A.G.; Lamanuzzi, A.; Ria, R.; Storlazzi, C.T.; Mariggiò, M.A.; Vacca, A.; Frassanito, M.A. Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple Myeloma. Cells 2020, 9, 167. [Google Scholar] [CrossRef]
  29. Loda, S.; Krebs, J.; Danhof, S.; Schreder, M.; Solimando, A.G.; Strifler, S.; Rasche, L.; Kortüm, M.; Kerscher, A.; Knop, S.; et al. Exploration of Artificial Intelligence Use with ARIES in Multiple Myeloma Research. J. Clin. Med. 2019, 8, 999. [Google Scholar] [CrossRef] [Green Version]
  30. Massaro, A.; Galiano, A.; Scarafile, D.; Vacca, A.; Frassanito, A.; Melaccio, A.; Solimando, A.; Ria, R.; Calamita, G.; Bonomo, M.; et al. Telemedicine DSS-AI multi level platform for monoclonal gammopathy assistance. In Proceedings of the 2020 IEEE International Symposium on Medical Measurements and Applications (MeMeA), Bari, Italy, 1 June–1 July 2020; IEEE: Bari, Italy, 2020; pp. 1–5. [Google Scholar]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Solimando, A.G.; Krebs, M.; Bittrich, M.; Einsele, H. The Urgent Need for Precision Medicine in Cancer and Its Microenvironment: The Paradigmatic Case of Multiple Myeloma. J. Clin. Med. 2022, 11, 5461. https://doi.org/10.3390/jcm11185461

AMA Style

Solimando AG, Krebs M, Bittrich M, Einsele H. The Urgent Need for Precision Medicine in Cancer and Its Microenvironment: The Paradigmatic Case of Multiple Myeloma. Journal of Clinical Medicine. 2022; 11(18):5461. https://doi.org/10.3390/jcm11185461

Chicago/Turabian Style

Solimando, Antonio Giovanni, Markus Krebs, Max Bittrich, and Hermann Einsele. 2022. "The Urgent Need for Precision Medicine in Cancer and Its Microenvironment: The Paradigmatic Case of Multiple Myeloma" Journal of Clinical Medicine 11, no. 18: 5461. https://doi.org/10.3390/jcm11185461

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop