Next Article in Journal
A Fused Deep Learning Architecture for the Detection of the Relationship between the Mandibular Third Molar and the Mandibular Canal
Previous Article in Journal
Retinal and Choroidal Thinning—A Predictor of Coronary Artery Occlusion?
Previous Article in Special Issue
Comprehensive Development and Implementation of Good Laboratory Practice for NGS Based Targeted Panel on Solid Tumor FFPE Tissues in Diagnostics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue: Next-Generation Sequencing in Tumor Diagnosis and Treatment II

by
Umberto Malapelle
1,*,†,
Matteo Fassan
2,3,*,† and
Dario de Biase
4,*,†
1
Department of Public Health, University of Naples Federico II, 80138 Naples, Italy
2
Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy
3
Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy
4
Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2022, 12(8), 2017; https://doi.org/10.3390/diagnostics12082017
Submission received: 8 August 2022 / Revised: 18 August 2022 / Accepted: 19 August 2022 / Published: 20 August 2022
(This article belongs to the Special Issue Next-Generation Sequencing in Tumor Diagnosis and Treatment II)
Next-generation sequencing (NGS) allows for the sequencing of multiple genes at a very high depth of coverage. The principle of targeted therapy consists of the application of drugs targeted against well-defined molecules that play a key role in tumor progression and/or survival. Considering the continuous discovery of new molecules as a putative target or as being responsible for treatment resistance mechanisms, single-gene analyses are becoming less effective. At present, precision medicine requires multigene characterization. The introduction of NGS to molecular diagnostics has allowed us to combine the high analytical sensitivity with multigene tests [1,2,3]. The aim of this Special Issue is to focus on the application of NGS in characterizing molecular alterations in solid tumors for diagnostic, prognostic, or predictive purposes.
NGS provides a huge amount of molecular data starting from a relatively small amount of input material. However, this technique must not be considered a magician’s hat; the GIGO concept (Garbage In, Garbage Out), borrowed from computer science, is extremely valid also and above all for NGS analysis. Analyzing a total of 144 formalin-fixed and paraffin-embedded (FFPE) specimens, Chougule and colleagues performed a benchmark in quality check metrics of DNA and RNA input that should be utilized by molecular diagnostic laboratories for successful library preparation and good quality of NGS data [4]. The authors observed that samples with DIN (DNA integrity number) < 3 as well as DNA concentration < 5 ng/µL are likely to fail and that compensating the poor DNA quality with a higher DNA concentration is likely to give bad-quality NGS data. For FFPE RNA, they observed that RIN (RNA integrity number) is not an accurate quality indicator and that the RNA distribution value (DV) is a better-quality metric. Moreover, RNA library concentration is an important predictive parameter for successful RNA libraries [4]. As regards coverage and variant allele frequency cut-off for considering a “good-quality NGS sample”, the authors asserted that 250× coverage and 10% VAF (Variant Allele Frequency) have a high possibility of false-negative results and that coverage of 500× with 5% VAF is recommended for FFPE samples [4].
NGS panels would help also in characterizing the molecular alterations in clinically puzzling tumors. Malvi and colleagues have characterized a cohort of pancreatic ductal adenocarcinomas (PDAC) in order to find any molecular alterations that could be associated with histopathological features and clinical outcomes [5]. The authors have observed that the survival of patients with PDAC was related to the presence of the TP53 and/or KRAS mutation. In fact, patients with PDAC harboring the concomitant KRAS/TP53 mutations had a significantly worse OS if compared to those with PDAC harboring only one of the two genes mutated or without KRAS and TP53 mutations. Moreover, a dramatic prognostic difference in the KRAS/TP53 double-mutated PDAC patients with an N2 stage compared to all the other patients was observed [5]. NGS can then be an alternative technique to PCR-based assays if it is not associated with extra costs, as stated by the 2020 ESMO guidelines [6], to evaluate KRAS and TP53 molecular status in PDAC to improve the clinical management of the patients.
In mPDAC, the first-line therapy is based on chemotherapy (gemcitabine combined with nab-paclitaxel, or FOLFIRINOX) [7]. In 2019, the FDA (Food and Drug Administration) approved the use of PARP inhibitors (i.e., Olaparib) as maintenance for PDAC patients harboring germline BRCA1 or BRCA2 pathogenic mutations (gBRCAm) [8]. In this context, the assessment of BRCA1 and BRCA2 mutational status is nowadays crucial in PDAC patients. Bruno and colleagues evaluated the feasibility of BRCA1/2 testing by NGS panel on a series of FFPE pancreatic tumor clinical specimens [9]. Starting from a median input DNA of 10 nanograms, they observed that 86.5% of cases were adequate for NGS analysis, with a success rate of 81.2%. Intriguingly, the failed specimens were all from tissue macrosections, characterized by a higher rate of fragmented DNA than standard sections, biopsies, and fine-needle aspirations, due to the formalin fixation procedure [9].
Liquid biopsy refers to a minimally invasive method of analysis of molecular neoplastic biomarkers performed starting from any type of patient body fluid (e.g., plasma, bile, urine, saliva, cerebrospinal fluid, and pleural effusion) [10,11]. To date, liquid biopsy analysis is routinely used in clinical practice, mainly in lung lesions, for obtaining material for molecular analyses if the “solid tissue” material is suboptimal, monitoring the treatment response, and detecting the minimal residual disease [12].
Zulato and colleagues have implemented the use of Roche’s Avenio ctDNA expanded panel in about 90 diagnostic routine non-small cell lung cancers (NSCLCs) [13]. They were able to successfully sequence 96.5% of samples. Avenio ctDNA kits demonstrated 100% sensitivity in detecting single nucleotide variants (SNVs) at VAF higher than 0.5% and high consistency in reproducibility. Moreover, they obtained matched results between tissue and liquid samples in 89% of analyzed specimens [13].
Simarro and colleagues described a case report of a patient with metastatic NSCLC who developed resistance mechanisms to the first-line EGFR-TKI treatment (dacomitinib), and to the second-line treatment with Osimertinib [14]. NGS analysis performed in liquid biopsy to characterize the resistance mechanism to second-line treatment revealed the founder deletion in exon 19 of the EGFR gene in concomitancy with a TP53 deletion [14]. Further analysis allowed for the detection of a truncating mutation in the RB1 gene, providing solid evidence of the resistance mechanism to second-line treatment with Osimertinib [14]. This study highlights the importance of implementing these high throughput molecular techniques in routine clinical practice, to understand the genomic heterogeneity of the tumor leading to personalized molecular-guided treatment.
Liquid biopsy is a useful tool not only for the management of NSCLC. Kastrisiou and colleagues had developed a targeted, cost-effective NGS gene panel that could be easily integrated in the day-to-day clinical routine starting from the plasma of patients with metastatic colorectal carcinomas (mCRC) [15]. The panel allows for the analysis of the hotspots in six clinically mCRC-relevant genes (KRAS, NRAS, MET, BRAF, ERBB2, and EGFR) with a negative and positive agreement of RAS Testing in Tissue and Plasma specimens of 92.8% and 81.2%, respectively [15].
As reported above, NGS multi-gene panel may be useful also to better characterize clinically interesting cases. De Falco and colleagues described two patients with CRC at different stages (pT2N0M0 and pT4cN1cM1), but both of them harbored double concurrent KRAS pathogenic mutations (p.G12D and p.G13D) that are normally mutually exclusive [15]. Moreover, using an NGS panel, the authors observed that the two tumors also harbored other mutations in PIK3CA, SMAD4, NOTCH1, ERBB2, and EGFR genes, but all of them were different in the two tumors [15].
Catino and colleagues described the clinical outcome of a patient acquiring multiple ALK mutations after lorlatinib treatment. In the same initial sites and at the abdominal lymph nodes and contralateral pleura, the plasma of the patient was analyzed using a multi-gene panel that confirmed an EML4-ALK fusion variant and also revealed the presence of ALK mutation p.G1202R [16]. At disease progression after ALK-TKI lorlatinib treatment, another multi-gene NGS assay was performed on cell blocks of pleural effusions revealing that the ALK p.G1202R mutation was still present and that another ALK missense mutation (p.T1151K) was found. The authors conclude that this case emphasizes the need to retest patients during the disease course and at the time of progression during ALK-TKIs therapy [16].

Funding

This research received no external funding.

Conflicts of Interest

Umberto Malapelle reports personal fees (as speaker bureau or advisor) from Boehringer Ingelheim, AstraZeneca, Roche, M.S.D., Amgen, G.S.K., Merck, Diaceutics, Eli Lilly, Thermofisher, Diatech, Hedera, unrelated to the current work. Matteo Fassan has received research grants from Astellas Pharma, Macrophage Pharma and Q.E.D. Therapeutics and reports personal fees (as speaker bureau or advisor) from Astellas Pharma, M.S.D., Astra Zeneca, Piere Frabre, Roche, G.S.K., unrelated to the current work. Dario de Biase has received personal fees (as consultant and/or speaker bureau) from Boehringer Ingelheim, and Eli Lilly, unrelated to the current work.

References

  1. Cappello, F.; Angerilli, V.; Munari, G.; Ceccon, C.; Sabbadin, M.; Pagni, F.; Fusco, N.; Malapelle, U.; Fassan, M. FFPE-Based NGS Approaches into Clinical Practice: The Limits of Glory from a Pathologist Viewpoint. J. Pers. Med. 2022, 12, 750. [Google Scholar] [CrossRef] [PubMed]
  2. de Biase, D.; Acquaviva, G.; Visani, M.; Sanza, V.; Argento, C.M.; De Leo, A.; Maloberti, T.; Pession, A.; Tallini, G. Molecular Diagnostic of Solid Tumor Using a Next Generation Sequencing Custom-Designed Multi-Gene Panel. Diagnostics 2020, 10, 250. [Google Scholar] [CrossRef] [PubMed]
  3. Pisapia, P.; Pepe, F.; Baggi, A.; Barberis, M.; Galvano, A.; Gristina, V.; Mastrilli, F.; Novello, S.; Pagni, F.; Pasini, S.; et al. Next generation diagnostic algorithm in non-small cell lung cancer predictive molecular pathology: The KWAY Italian multicenter cost evaluation study. Crit. Rev. Oncol. Hematol. 2022, 169, 103525. [Google Scholar] [CrossRef] [PubMed]
  4. Chougule, A.; Jagtap, V.; Nikam, A.; Kale, S.; Nambiar, K.; Bagayatkar, P.; Chandrani, P.; Kaushal, R.; Noronha, V.; Patil, V.; et al. Comprehensive Development and Implementation of Good Laboratory Practice for NGS Based Targeted Panel on Solid Tumor FFPE Tissues in Diagnostics. Diagnostics 2022, 12, 1291. [Google Scholar] [CrossRef] [PubMed]
  5. Malvi, D.; Vasuri, F.; Maloberti, T.; Sanza, V.; De Leo, A.; Fornelli, A.; Masetti, M.; Benini, C.; Lombardi, R.; Offi, M.F.; et al. Molecular Characterization of Pancreatic Ductal Adenocarcinoma Using a Next-Generation Sequencing Custom-Designed Multigene Panel. Diagnostics 2022, 12, 1058. [Google Scholar] [CrossRef] [PubMed]
  6. Mosele, F.; Remon, J.; Mateo, J.; Westphalen, C.B.; Barlesi, F.; Lolkema, M.P.; Normanno, N.; Scarpa, A.; Robson, M.; Meric-Bernstam, F.; et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: A report from the ESMO Precision Medicine Working Group. Ann. Oncol. 2020, 31, 1491–1505. [Google Scholar] [CrossRef] [PubMed]
  7. Taieb, J.; Prager, G.W.; Melisi, D.; Westphalen, C.B.; D’Esquermes, N.; Ferreras, A.; Carrato, A.; Macarulla, T. First-line and second-line treatment of patients with metastatic pancreatic adenocarcinoma in routine clinical practice across Europe: A retrospective, observational chart review study. ESMO Open 2020, 5, e000587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. FDA Approves Olaparib for gBRCAm Metastatic Pancreatic Adenocarcinoma. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-olaparib-gbrcam-metastatic-pancreatic-adenocarcinoma (accessed on 3 August 2022).
  9. Bruno, R.; Sensi, E.; Lupi, C.; Giordano, M.; Bernardini, L.; Vivaldi, C.; Fornaro, L.; Vasile, E.; Campani, D.; Fontanini, G. Feasibility of BRCA1/2 Testing of Formalin-Fixed and Paraffin-Embedded Pancreatic Tumor Samples: A Consecutive Clinical Series. Diagnostics 2021, 11, 1046. [Google Scholar] [CrossRef] [PubMed]
  10. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef] [PubMed]
  11. Malapelle, U.; Muscarella, L.A.; Pisapia, P.; Rossi, A. Targeting emerging molecular alterations in the treatment of non-small cell lung cancer: Current challenges and the way forward. Expert Opin. Investig. Drugs 2020, 29, 363–372. [Google Scholar] [CrossRef] [PubMed]
  12. de Biase, D.; Maloberti, T.; De Leo, A.; Sanza, V.; Visani, M.; Gruppioni, E.; Altimari, A.; Grillini, A.; Gelsomino, F.; Ferrari, M.; et al. Next-generation technologies in predictive molecular pathology of lung cancers. J. Xiangya Med. 2022, 7, 15. [Google Scholar] [CrossRef]
  13. Zulato, E.; Tosello, V.; Nardo, G.; Bonanno, L.; Del Bianco, P.; Indraccolo, S. Implementation of Next Generation Sequencing-Based Liquid Biopsy for Clinical Molecular Diagnostics in Non-Small Cell Lung Cancer (NSCLC) Patients. Diagnostics 2021, 11, 1468. [Google Scholar] [CrossRef] [PubMed]
  14. Simarro, J.; Perez-Simo, G.; Mancheno, N.; Munoz-Nunez, C.F.; Cases, E.; Juan, O.; Palanca, S. Utility of Next-Generation Sequencing in the Reconstruction of Clonal Architecture in a Patient with an EGFR Mutated Advanced Non-Small Cell Lung Cancer: A Case Report. Diagnostics 2022, 12, 1266. [Google Scholar] [CrossRef] [PubMed]
  15. Kastrisiou, M.; Zarkavelis, G.; Kougioumtzi, A.; Sakaloglou, P.; Kostoulas, C.; Georgiou, I.; Batistatou, A.; Pentheroudakis, G.; Magklara, A. Development and Validation of a Targeted ‘Liquid’ NGS Panel for Treatment Customization in Patients with Metastatic Colorectal Cancer. Diagnostics 2021, 11, 2375. [Google Scholar] [CrossRef] [PubMed]
  16. Catino, A.; Lacalamita, R.; De Summa, S.; Pesola, F.; Tommasi, S.; Galetta, D. Multiple Genetic Alterations as Resistance Mechanism during Second-Line Lorlatinib for Advanced ALK-Rearranged Lung Adenocarcinoma: A Case Report. Diagnostics 2022, 12, 682. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Malapelle, U.; Fassan, M.; de Biase, D. Special Issue: Next-Generation Sequencing in Tumor Diagnosis and Treatment II. Diagnostics 2022, 12, 2017. https://doi.org/10.3390/diagnostics12082017

AMA Style

Malapelle U, Fassan M, de Biase D. Special Issue: Next-Generation Sequencing in Tumor Diagnosis and Treatment II. Diagnostics. 2022; 12(8):2017. https://doi.org/10.3390/diagnostics12082017

Chicago/Turabian Style

Malapelle, Umberto, Matteo Fassan, and Dario de Biase. 2022. "Special Issue: Next-Generation Sequencing in Tumor Diagnosis and Treatment II" Diagnostics 12, no. 8: 2017. https://doi.org/10.3390/diagnostics12082017

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