Get Permission Patel, Gunjan, and Vanteddu: Bio-markers of immuno-oncology


Introduction

The idea(s) behind immuno-(I-O) oncology's possible uses have existed for a while. The first attempts to use heat-inactivated Streptococcus pyogenes and Serratia marcescens to treat osteosarcoma were made more than a century ago, in 1891, by the early pioneer William Coley. This marked the beginning of research into using immune pathways to treat cancer.1, 2 The identification of programmed cell death protein 1 (PD-1) by Honjo and colleagues in the 2000s and cytotoxic T-lymphocyte antigen 4 (CTLA-4) by Allison and colleagues in the 1990s led to the development of a new class of anticancer medications known as immune checkpoint inhibitors (ICIs). ICIs are currently widely used to treat many kinds of cancer. 2

The oncology community's attitude on I-O therapies has evolved over time as a result of multiple breakthroughs, challenges, and attempts to attain therapeutic success. The concept of tumor-infiltrating lymphocytes (TILs) and the development of effector T cells that express chimeric antigen receptors (CAR T) are two examples of advancement. Two instances of encouraging preclinical data that are occasionally followed by failures in subsequent clinical trials include the failures of I-O combination therapy in several tumor types. This intriguing therapeutic strategy has recently become a reliable clinical reality, which has caused the I-O field to quickly gather speed. 3, 4

Biopharmaceutical companies have made large investments in the I-O region due to its rapid advances and practice-altering clinical victories for many difficult-to-treat tumors. Additionally, the area has inspired active research into new preclinical models and biomarkers to better our understanding. 5 To comprehend improved predictive preclinical models and translational methodologies are now more critical than ever. Furthermore, if current problems, disagreements, and constraints are resolved in the completed and ongoing clinical trials of ICIs, it will be feasible to continue giving patients new I-O medications. 6, 7

The three main elements that can improve the effectiveness of drug development are identifying the right biomarkers to select the right patient population, recommending the engagement of the relevant target, and carrying out the formal clinical planning procedure. 8

Discussion

Immuno-oncology non-clinical research models

Preclinical oncology studies come in various forms, each with unique benefits and drawbacks, including tumor syngeneic mouse animal models in-vivo, genetically modified mouse models (GEMMs), patient-derived xenograft, mouse humanized models, and onco organoids/spheroids. Researchers must consider every model's characteristic in the content of this particular research queries when deciding which model is most appropriate.9, 10

More than the years, the Immuno-Oncology configuration has been the most frequently utilized for synthetic mouse models. To create these models, existing cell lines belong to mouse or tumor engrafts are transplanted into immune-competent, strain-matched mouse hosts. 11 Commercially available syngeneic mouse models for hematological and solid tumors include ones engineered to consistently amplification of the luciferase gene to enable the utilization of in vivo animal imaging. 12, 13

These systems are simple to use logistically, can screen a huge number of drug candidates, and can be used to explore pharmacodynamics and the mechanism of action. Syngeneic models have limitations despite their widespread use and relevance to human tumors. 14, 15 Since the tumor cells are originated from mice, for instance, not all characteristics of clinical disease in humans can be accurately predicted by the histocytes of mouse tumors. Additionally, these models' predictions of clinical outcomes for treatment responses can be subpar. 16 However, many of these drawbacks are outweighed by their use in understanding crucial elements of immunological response to therapy. 17, 18

Genetically altered, transgenic mouse models, also known as GEMMs, have been found to more closely monitor the affecting the supporting tissue biology of their human disease equivalents than syngeneic models, includes the tumor biology, which has a significant impact on tumor activity. 19, 20 With the help of tissue-targeted factors and applicable mutations that are known to encourage tumor stimulation in the normal tissue or organ of specified, these animal models enable a more natural process of disease initiation. Historically, these models have been crucial for developing oncology drugs and offered insightful information regarding carcinogenesis. 21 For instance, Dow et al. have demonstrated the validation of therapeutics targeting the Wnt signaling pathway in colorectal cancer using a transgenic model.22, 23

Although certain GEMMs and syngeneic mouse cell line models are helpful in developing I-O medicines, CDX models for human cancer must also be considered.24 These also have the industries level for researching cell-based immunotherapies, even though they have been actively utilized for more traditional oncology techniques. With thousands of commercially accessible human cell lines, many of which stably express the firefly luciferase gene, CDX models are logistically simple to utilize, making them excellent for in vivo small animal imaging. 25, 26, 27 To explore the functions of the immune system in tumor formation and therapy response, these investigations must be carried out in immune-deficient mice strains.28, 29

To address some of the drawbacks of conventional CDX animal models, PDX mouse animal models, depending on transplants and in-vivo repeated dissemination of modern human tumor biopsies in immune-deficient mice, has been created. 30 Even after recurrent passaging in mice, unlike CDX tumors, Patient Derived Xenograft tumors retain the genomic, chromosomal, and morphological changes seen in cancer patients, making them superior projections of clinical prognosis in human beings.31, 32 Additionally, Patient Derived Xenograft tumors are frequently defined, allowing for more detailed analyses of drug resistance and therapeutic uses. There are a few well-known problems with mentioned animal models, though. Although thousands of these cell-lines exist, some more tumor histology like prostate tumor and several hematology tumors—are much more challenges to identify.33, 34, 35

The main drawbacks of syngeneic and GEMMs are the absence of human targets and the innate immunologic variations between animals. As a result, a lot of work has gone toward humanizing the mouse immune system. 36 The immunoavatar animal model, which involves regenerating immune-deficient mice with human type of peripheral blood mononuclear cells (PBMCs), is one of the more popular methods for humanizing mouse models. 37, 38, 39 Even though complex purification methods are not required, substantial human xenograft-versus-host illness.40, 41

As an alternative, it has been shown that immunocompromised-NOD SCID gamma (or NSG) mice results in the development of numerous lineages of human hematopoietic and immunological cells. 42 The immune systems of these so-called HuNSG mice eventually become somewhat functional. It has become a busy area of research thanks to the expansion of humanized mice models, which have made it possible to define human cell function of CAR T in vivo and simplify the creation of new development of CAR therapeutics. 43 Mostly of this, each and every humanly optimized mouse method has particular benefits and drawbacks that should be examined before usage.42, 43

Organoid technology has recently been used to produce a unique cell-based method for studying tumor immunobiology that is swiftly being adjusted to the I-O and environment. Organoids are three-dimensional in vitro cultures of healthy or malignant tissues that contain a variety of cell lineages, such as stem cells and differentiated cells, as well as tissue architecture. Organoids holds potential as a more clinically significant preclinical theoretical model for drug development and precision medicine than cell lines because of their closeness to the original malignancies and the capacity to culture organoids from each patient. Oncology and I-O medicines are being discovered and developed using various in vitro or ex vivo techniques. 44

In conclusion, the proliferation of I-O clinical investigations has sparked new research into mice with functioning immune systems. As a result, the variety of tumor histotypes represented in syngeneic models is quickly increasing.45 Syngeneic models with malignancies developed in orthotopic environments are also becoming more popular. This makes it possible to surgically insert tumor cells right into the organ, track and monitor their growth, and assess how well the treatment works. Using luciferase-expressing cell lines, transversal, non-invasive small animal bioluminescence imaging can be used in these situations, drastically reducing the number of mice used in the study and enabling semi-quantitative measurement of tumor burden. 45, 46

Synthetic mouse models can be used to find therapy response biomarkers

According to reports, synthetic mice models react variably to ICIs. This responsive is established for most of frequently employed model.47, 48 These models differ in the general elements of the tumor micro environment, in addition to changes in therapy response. Because of these variations, many of the models have been categorized where cold animal models tend to have a infiltration of both.49

To identify the most appropriate model for preclinical research, it is necessary to characterize the baseline tumor using the following metrics: (a) the infiltrating leukocyte population; (b) other molecular features of the models (RNASeq, whole exome sequencing, etc.); and (c) benchmark therapy efficacy; and to use these baseline treatment responses to study drug usages and identify phenotypic indicators of response.50 For instance, in the 4T1-luciferase type of mouse mammary cancer, an immunologically "cold" tumor, the initial cancer is known to spread to the neck area, includes the lung and lymph nodes (axillary). 51

Compared to anti-CTLA-4 or focused in this model, marginally shrinks the primary tumor size; however, it significantly delays the emergence of metastatic illness. Bioluminescence imaging techniques can be used to see, measure, and compare these therapy responses. 52 Furthermore, found and measured using flow cytometry and serial blood samples. 53

Additionally, biomarkers can be utilized to choose the best models of researching a specific I-O agent or spot gene expression changing after treatment. For instance, microarray data demonstrate that successfully treated with immuno-modulatory agents, exhibit very different gene expression patterns, and this knowledge can be used to identify gene response. 54

Clinical use of predictive biomarkers in immuno-oncology medication development

Biomarkers help evaluate many aspects of treatment response in preclinical models, as was previously indicated, but they are also increasingly used in the clinic for patient management. Biomarkers are "specified characteristics that are tested as indicators of normal biochemical functions, pathogenic processes, or reactions to an exposure or treatment, particularly treatment modalities," according to the US Food and Drug Administration (FDA). 55 Various biomarkers, including diagnostic, prognostic, and predictive biomarkers, are utilized to guide therapy decisions to maximize patients' reactions to different treatments. Predictive biomarkers are used to determine the likelihood of a clinical outcome, such as a disease recurrence or progression. 56 At the same time, diagnosing indicators are employed to identify or prove the presence of a disease or condition of interest. 57, 58

Clinical trials frequently use prognostic indicators to establish trial inclusion criteria to pinpoint highly-risk of the patients. The term "predictive biomarkers" refers to a changing in a biomarker this indicates whether a specific or group of particulars is more to experience a positive or negative reaction to exposure of specific medicinal agent; these biomarkers are particularly helpful in the planning and execution of clinical trial. 59 The classification of some diseases may change due to the development of diagnostic biomarkers. 60 Many predictive bio therapeutic agents are employed in diagnostic to maximize patients' reactions to different therapies, and others are used to advise therapy choices. 61, 62

Immunohistochemistry (IHC) staining is therefore advised prior to beginning a number of such therapies, such as pembrolizumab, which utilizes the PD-L1 SP142 IHC assay established by studies have demonstrated that patients with high TMBs have longer projection-free survive and length of responsiveness to onco drugs than patients with lower mutational burdens. 63

Research is now being done to establish the threshold values at the place of tumor cell mutation is deemed high, and to create practical diagnostic cellular assays and techniques of simplification. Predictive biomarkers are so crucial for guiding therapeutic agents that many tumor treatments are now available based on the main biomarkers linked with the tumor rather than where cancer started in the body. The immunosurveillance system's ability is improved due to TMB's promotion of the synthesis of neoantigens expressed on MHC. 64

The fact that ICIs primed with chemotherapy are more effective against BRCA1-mutated TNBC cancers elevated TMB suggests that clinically determining BRCA1 status could act as a prognostic therapeutic agent. Interestingly, highest TMB (the overall mutants to the actual quantity of sequential mega base assessed using MSK-IMPACT (assay)) is associated with greater survival rates. This was discovered by studying the new genetic and medical evidence of 1662 advanced cancer patients treated with FDA-approved ICIs. 65 for the patient, this means that while deciding on a final course of treatment, it is crucial to concentrate efforts on malignant cells biochemical pathways that drive cancer growth and pinpoint the precise origin of their malignancies. 66

Biomarkers primarily been investigated independently until recently. Still, current invention is focusing toward examining the variations of bioagents that address characteristics of the cancer, which include gene expression are tried to apply to clinical response and patient care. Such methods might increase the success of clinical trials for cutting-edge treatments or combinations. 67 To evaluate this crucial interaction, describe the patient response, and assist reverse translational techniques, is a helpful strategy. Tumor inflammation signatures, signatures associated with CD8-positive cells invading tumors, and signatures that combine the indicators are only a few of the gene expression signatures that have been established. 68

Seven different types of biomarkers were detailed in the "Biomarkers, endpoints, and other Tools" framework, published on 2016 by the FDA. The monitoring, highly susceptible, safety, and pharmacodynamics biomarkers were also defined in this framework in add to clinical, predictive, and respective biomarkers covered. 69

Paradigm for clinical trials is changing due to immuno-oncology

The general strategy for systemic anticancer therapy has changed during the past few decades. Chemicals used in cytotoxic chemotherapy were directed at any dividing cell. Then came targeted, which functioned only on cancer cells with particular molecular abnormalities. Recent advances in immunotherapy can be broadly divided into two categories: those that directly affect cancer cells (known as passive) and encourage immunological cells (known as active). 60, 61

The genomic characteristics of many tumors are becoming better understood as high-throughput techniques like next-generation sequencing become more widely available and less expensive. As a result, cancer patients are being divided into more manageable subpopulations, and populations with positive biomarkers are now enrolled in Immuno-Oncology trials based on the unique molecular level features of malignancies. 62, 63

A dramatic decrease in the time needed of clinical target of novel therapeutic anticancer medicines. For cytotoxicological agents, the new drug development procedures entailed a series of trials: from phase I to establish therapeutic dose and human safety, and phase II to fix the therapeutic dose, record unwanted effects, and prove the efficacy, phase III to ensure therapeutic effect and evaluate the new cancer treatment in comparison to the industry standard. 64 Regulatory approval was then given based on the positive outcomes of phase III. In the era of targeted medicines, encouraging phase II results occasionally sufficed to obtain accelerated approval, subject to the completion of subsequent confirmatory phase III studies. In the current era of immunotherapy, development frequently starts with a modest phase I study, with the gradual addition of various extensions cohorts for the same research, perhaps with randomized arms. 65, 66

One treatment may be used for several most diseases (a basket), many treatments may be applied to a single illness (an "umbrella study") with the use of "master" protocols. 67

The Kaplan-Meier curves (I-O vs. standard chemotherapy) have an exciting tendency to separate later but eventually lead to a sustained separation (often evocatively referred to as "raising the tail of the curve"), signifying a durable advantage found in a percentage of patients. From a statistical perspective, this presents methodological difficulties. Therefore, in addition to progression-free survival, other more recent approaches, including landmark survival, landmark analysis, and confined mean survival rate, is additionally being researched in the I-O era. 68, 69, 70

The oncology discipline has seen a substantial transition toward precision medicine concurrent with the emergence of I-O compounds. Precision medicine uses data on a person's cancer-related characteristics to identify and treat the specific disease. 70

Conclusion

Increasing I-O therapy clinical trials have sparked new studies using immune system-competent mouse models. More thorough preclinical trials are required participation and identify biomarkers explored during further stages of medication invention. A more concentrated effort is being made the correct volunteers, which is also causing significant changes in the methodology, design, and implementation of cancer clinical trials. Biomarkers are now a crucial component of the improvement to achieve this patient population.

Pembrolizumab illustrates a drug approval based on a phase I expansion study with patient-driven biomarkers. Populations refer to this novel approach to clinical development that requires fewer individuals and significantly reduces the time to approval. There is now the use of new technology, such as liquid biopsies.

This new technology will make it simpler for patients to continue taking part in further I-O studies because it has recently been approved as a diagnostic technique for NSCLC and breast cancer. Drug development cycles are getting shorter thanks to the trend towards smaller precision trials determined by biomarkers, which is also opening the door for more adaptable clinical trial models.

Indeed, the FDA's recent clearance of cancer indications based only on patient biomarkers offers hope for the future48, but newer, more adaptable models still need to be thoroughly tested. Even though preclinical models have contributed a significant amount of knowledge to aid in the clinical development of ICIs, further improvement of these models is encouraged to more accurately mimic human cancer.

Preclinical and clinical development needs to be more closely linked as treating various tumors with these treatments, becomes increasingly important. Linking non-clinical and clinical development requires using biotherapeutic agents that can aid in response prediction or provide insight into the nature of the treatment.

The prospective success rates of innovative therapeutics and immuno-oncology combo medicines will increase due to new drug inventions made possible by translation and reverse translational methodologies.

Source of Funding

None.

Conflict of Interest

None.

References

1 

P Dobosz T Dzieciątkowski The intriguing history of cancer immunotherapyFront Immunol2019102965

2 

RD Carlson JC Flickinger AE Snook Talkin’ toxins: from Coley’s to modern cancer immunotherapyToxins202012241

3 

JA Gross T St John JP Allison The murine homologue of the T lymphocyte antigen CD28. Molecular cloning and cell surface expressionJ Immunol Baltim19901448320111

4 

T Okazaki Y Iwai T Honjo New regulatory co-receptors: inducible co-stimulator and PD-1Curr Opin Immunol200214677982

5 

JJ Adashek S Kato R Ferrara Hyperprogression and immune checkpoint inhibitors: hype or progress?Oncologist202025294102

6 

RM Frederickson A new era of innovation for CAR T-cell therapyMol Ther201523121795801

7 

KS Saini IM Svane M Juan Manufacture of adoptive cell therapies at academic cancer centers: scientific, safety and regulatory challengesAnn Oncol2021331612

8 

N Tokarew J Ogonek S Endres Teaching an old dog new tricks: next-generation CAR T cellsBr J Cancer20191202637

9 

CR Parish Cancer immunotherapy: the past, the present and the futureImmunol Cell Biol20038110619

10 

M Kocikowski K Dziubek M Parys Hyperprogression under immune checkpoint-based immunotherapy-current understanding, the role of PD-L1 tumour-intrinsic signalling, future directions and a potential large animal modelCancers202012804

11 

S Dudani J Graham JC Wells First-line immuno-oncology combination therapies in metastatic renal-cell carcinoma: results from the International metastatic renal-cell carcinoma database consortiumEur Urol20197668618

12 

C Robert L Thomas I Bondarenko Ipilimumab plus dacarbazine for previously untreated metastatic melanomaN Engl J Med2011364251726

13 

MD Hellmann L Paz-Ares B Caro Nivolumab plus ipilimumab in advanced non-small-cell lung cancerN Engl J Med201938121202051

14 

S Upadhaya VM Hubbard-Lucey JX Yu Immuno-oncology drug development forges on despite COVID-19Nat Rev Drug Discov2020197513

15 

CS Baik EH Rubin PM Forde Immuno-oncology clinical trial design: limitations, challenges, and opportunitiesClin Cancer Res2017231749925002

16 

M Smoragiewicz J Bogaerts E Calvo Design and conduct of early clinical studies of immunotherapy agent combinations: recommendations from the task force on methodology for the development of innovative cancer therapiesAnn Oncol20182911217582

17 

B Olson Y Li Y Lin Mouse models for cancer immunotherapy researchCancer Discov2018811135865

18 

M He M Henderson S Muth Preclinical mouse models for immunotherapeutic and non-immunotherapeutic drug development for pancreatic ductal adenocarcinomaAnn Pancreat Cancer202037122

19 

W Zhong JS Myers F Wang Comparison of the molecular and cellular phenotypes of common mouse syngeneic models with human tumorsBMC Genomics20202122 10.1186/s12864-019-6344-3

20 

J Murphy Pre-clinical murine models: syngeneic models for immuno-oncologyMOJ Immunol20152452

21 

P Rampetsreiter E Casanova R Eferl Genetically modified mouse models of cancer invasion and metastasisDrug Discov Today201182-36774

22 

K Kersten KE Visser MH Van Miltenburg Genetically engineered mouse models in oncology research and cancer medicineEMBO Mol Med20179213753

23 

LE Dow KP O’rourke J Simon Apc restoration promotes cellular differentiation and reestablishes crypt homeostasis in colorectal cancerCell20151617153952

24 

A Goodspeed LM Heiser JW Gray Tumor-derived cell lines as molecular models of cancer pharmacogenomicsMol Cancer Res2016141313

25 

VC Daniel L Marchionni JS Hierman A primary xenograft model of small-cell lung cancer reveals irreversible changes in gene expression imposed by culture in vitroCancer Res2009698336473

26 

JP Gillet AM Calcagno S Varma Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistanceProc Natl Acad Sci USA2011108461870821

27 

J Noorbakhsh F Vazquez JM Mcfarland Bridging the gap between cancer cell line models and tumours using gene expression dataBr J Cancer20211253113

28 

V Vidhyasagar SU Haq BH Lok Patient-derived xenograft models of small cell lung cancer for therapeutic developmentClin Oncol2020321061944

29 

LD Shultz MA Brehm JV Garcia-Martinez Humanized mice for immune system investigation: progress, promise and challengesNat Rev Immunol2012121178698

30 

F Weeber SN Ooft KK Dijkstra Tumor organoids as a pre-clinical cancer model for drug discoveryCell Chem Biol20172491092100

31 

J Rodrigues MA Heinrich LM Teixeira In vitro model (R) evolution: unveiling tumor-stroma interactionsTrends Cancer20217324964

32 

DS Chulpanova KV Kitaeva CS Rutland Mouse tumor models for advanced cancer immunotherapyInt J Mol Sci20202111411810.3390/ijms21114118

33 

MA Taylor AM Hughes J Walton Longitudinal immune characterization of syngeneic tumor models to enable model selection for immune oncology drug discoveryJ Immunother Cancer20197132810.1186/s40425-019-0794-7

34 

JW Yu S Bhattacharya N Yanamandra Tumor-immune profiling of murine syngeneic tumor models as a framework to guide mechanistic studies and predict therapy response in distinct tumor microenvironmentsPLoS One2018131120622310.1371/journal.pone.0206223

35 

JJ Arroyo-Crespo A Armiñán D Charbonnier Characterization of triple-negative breast cancer preclinical models provides functional evidence of metastatic progressionInt J Cancer20191458226781

36 

S Demaria N Kawashima AM Yang Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancerClin Cancer Res Off J Am Assoc Cancer Res200511172862

37 

MR Franklin Preclinical assessment of anti-tumor activity and immune response in syngeneic tumor models (Poster 294) the 28th EORTC-NCI-AACR Symposium2016https://biopharma.labcorp.com/industry-solutions/by-therapeutic-area/oncology/preclinical/posters/preclinical-assessment-anti-tumor-activity-immune-response-syngeneic-tumor-models.html

39 

RM Califf Biomarker definitions and their applicationsExp Biol Med2018243321334

40 

R Bai Z Lv D Xu Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitorsBiomark Res202083410.1186/s40364-020-00209-0

41 

SP Patel R Kurzrock PD-L1 expression as a predictive biomarker in cancer immunotherapyMol Cancer Ther201514484756

42 

P Zhao L Li X Jiang Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacyJ Hematol Oncol20191215410.1186/s13045-019-0738-1

43 

F Marliot X Chen A Kirilovsky T Sbarrato C El Sissy L Batista Analytical validation of the immunoscore and its associated prognostic value in patients with colon cancerJ Immunother Cancer202081e00027210.1136/jitc-2019-000272

44 

GS Krigsfeld EA Prince J Pratt Analysis of real-world PD-L1 IHC 28-8 and 22C3 pharmDx assay utilisation, turnaround times and analytical concordance across multiple tumour typesJ Clin Pathol2020731065664

45 

B Vennapusa B Baker M Kowanetz Development of a PD-L1 complementary diagnostic immunohistochemistry assay (SP142) for AtezolizumabAppl Immunohistochem Mol Morphol201927292100

46 

JD Fumet C Truntzer M Yarchoan Tumour mutational burden as a biomarker for immunotherapy: current data and emerging conceptsEur J Cancer20201314050

47 

MD Hellmann T-E Ciuleanu A Pluzanski Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burdenN Engl J Med20183782093104

48 

KS Saini K Punie C Twelves Antibody-drug conjugates, immune-checkpoint inhibitors, and their combination in breast cancer therapeuticsExpert Opin Biol Ther202121794562

49 

L Fancello S Gandini PG Pelicci Tumor mutational burden quantification from targeted gene panels: major advancements and challengesJ Immunother Cancer20197118310.1186/s40425-019-0647-4

50 

TA Chan M Yarchoan E Jaffee Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinicAnn Oncol20193014456

51 

S Arora R Velichinskii RW Lesh Existing and emerging biomarkers for immune checkpoint immunotherapy in solid tumorsAdv Ther20193610263878

52 

K Kim AD Skora Z Li Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cellsProc Natl Acad Sci USA2014111321177483

53 

E Nolan P Savas AN Policheni Combined immune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancerSci Transl Med20179393

54 

RM Samstein CH Lee AN Shoushtari Tumor mutational load predicts survival after immunotherapy across multiple cancer typesNat Genet20195122028

55 

A Bhattacharyya SN Rai Adaptive signature design- review of the biomarker guided adaptive phase -III controlled designContemp Clin Trials Commun201915100378

56 

SK Singhal N Usmani S Michiels Towards understanding the breast cancer epigenome: a comparison of genome-wide DNA methylation and gene expression dataOncotarget201673300219

57 

S Dedeurwaerder C Desmedt E Calonne DNA methylation profiling reveals a predominant immune component in breast cancersEMBO Mol Med201131272667

58 

D Zardavas M Maetens A Irrthum The AURORA initiative for metastatic breast cancerBr J Cancer20141111018818

59 

C Criscitiello D Fumagalli KS Saini Tamoxifen in early-stage estrogen receptor-positive breast cancer: overview of clinical use and molecular biomarkers for patient selectionOnco Targets Ther2010411110.2147/OTT.S10155

60 

D Damotte S Warren J Arrondeau The tumor inflammation signature (TIS) is associated with anti-PD-1 treatment benefit in the CERTIM pan-cancer cohortJ Transl Med201917135710.1186/s12967-019-2100-3

61 

PM Szabo S Pant S Ely Development and performance of a CD8 gene signature for characterizing inflammation in the tumor microenvironment across multiple tumor typesJ Mol Diagn2021239115973

62 

FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource2016http://www.ncbi.nlm.nih.gov/books/NBK326791/

63 

C Criscitiello OM Filho KS Saini Targeted therapies in breast cancer: are heart and vessels also being targeted?Breast Cancer Res2012143209

64 

L Galluzzi E Vacchelli JM Bravo-San Pedro Classification of current anticancer immunotherapiesOncotarget201452412472508

65 

H Weinmann Cancer immunotherapy: selected targets and small-molecule modulatorsChem Med Chem201611545066

66 

JM Mehnert AM Monjazeb JMT Beerthuijzen The challenge for development of valuable immuno-oncology biomarkersClin Cancer Res2017231749709

67 

KT Flaherty R Gray A Chen The molecular analysis for therapy choice (NCI-MATCH) trial: lessons for genomic trial designJ Natl Cancer Inst2020112102130

68 

X Yu JH Lucey VM Tang Immuno-oncology drug development goes globalNat Rev Drug Discov20191812899900

69 

VT Devita E Chu A history of cancer chemotherapyCancer Res200868864353

70 

CA Umscheid DJ Margolis CE Grossman Key concepts of clinical trials: a narrative reviewPostgrad Med20111235194204



jats-html.xsl


This is an Open Access (OA) journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

  • Article highlights
  • Article tables
  • Article images

Article History

Received : 31-10-2023

Accepted : 10-01-2024


View Article

PDF File   Full Text Article


Copyright permission

Get article permission for commercial use

Downlaod

PDF File   XML File   ePub File


Digital Object Identifier (DOI)

Article DOI

https://doi.org/10.18231/j.jpbs.2023.017


Article Metrics






Article Access statistics

Viewed: 660

PDF Downloaded: 172



Medical Abbreviation List