Next Article in Journal
Selenium-Binding Protein 1 (SBP1): A New Putative Player of Stress Sensing in Plants
Previous Article in Journal
Olfactory Projections to Locomotor Control Centers in the Sea Lamprey
Previous Article in Special Issue
SLCO1B1 Exome Sequencing and Statin Treatment Response in 64,000 UK Biobank Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Receptor Pharmacogenomics: Deciphering Genetic Influence on Drug Response

by
Sorina Andreea Anghel
1,2,
Cristina-Elena Dinu-Pirvu
1,3,*,
Mihaela-Andreea Costache
1,
Ana Maria Voiculescu
1,
Mihaela Violeta Ghica
1,3,
Valentina Anuța
1,3 and
Lăcrămioara Popa
1,3
1
Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, 6 Traian Vuia Str., 020956 Bucharest, Romania
2
Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independentei 296, 060031 Bucharest, Romania
3
Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(17), 9371; https://doi.org/10.3390/ijms25179371
Submission received: 2 August 2024 / Revised: 23 August 2024 / Accepted: 26 August 2024 / Published: 29 August 2024
(This article belongs to the Special Issue Pharmacogenomics, 3rd Edition)

Abstract

:
The paradigm “one drug fits all” or “one dose fits all” will soon be challenged by pharmacogenetics research and application. Drug response—efficacy or safety—depends on interindividual variability. The current clinical practice does not include genetic screening as a routine procedure and does not account for genetic variation. Patients with the same illness receive the same treatment, yielding different responses. Integrating pharmacogenomics in therapy would provide critical information about how a patient will respond to a certain drug. Worldwide, great efforts are being made to achieve a personalized therapy-based approach. Nevertheless, a global harmonized guideline is still needed. Plasma membrane proteins, like receptor tyrosine kinase (RTK) and G protein-coupled receptors (GPCRs), are ubiquitously expressed, being involved in a diverse array of physiopathological processes. Over 30% of drugs approved by the FDA target GPCRs, reflecting the importance of assessing the genetic variability among individuals who are treated with these drugs. Pharmacogenomics of transmembrane protein receptors is a dynamic field with profound implications for precision medicine. Understanding genetic variations in these receptors provides a framework for optimizing drug therapies, minimizing adverse reactions, and advancing the paradigm of personalized healthcare.

1. Introduction

Over twenty years ago, the Human Genome Project, which provided the human genetic blueprint, was completed [1]. The genetic makeup of humans is 99.9% identical, while 0.1% accounts for genetic diversity resulting in phenotypic differences between individuals [2]. Each human being has a distinctive pattern of genetic variation that can impact disease susceptibility and drug response. The traditional medicine dogma is “one drug fits all” or “one dose fits all”, offering identical treatment to all patients that suffer from the same disease. However, the dosage can be individualized based on age or different comorbidities [3]. Using this approach, a great heterogeneity can be observed in drug response, giving rise to adverse reactions or treatment failure. In recent years, a more patient-focused type of medicine has surfaced, namely precision medicine. Modern tools such as technology-based omics, artificial intelligence, and big data analytics provide the interface that can depict individual variability and treat patients based on their uniqueness [4]. Pharmacogenetics and pharmacogenomics (PGx) are the pillars of precision medicine, attuning drug selection and dosage to each patient’s genetic profile [5]. Nevertheless, PGx testing is not a routine procedure. Since 2020, the U.S. Food and Drug Administration (FDA) issued a PGx information table that currently contains over 100 drug–biomarker pairs [6]. Moreover, for the last two decades, a powerful public database, PharmGKB (Pharmacogenomics Knowledgebase), has provided labeling information, clinical annotations, literature resources, and the pathways involved in gene–drug interactions [7,8,9]. Launched in 2009, the Clinical Pharmacogenetic Implementation Consortium (CPIC) has established clinical practice guidelines for PGx data, occasionally contrasting FDA drug labeling [10]. Thus, a global harmonized guideline needs to be implemented to translate PGx into clinical practice, increase the education of healthcare professionals, and offer patients personalized treatment.
PGx investigates the genetic variability in drug target genes (membrane receptors, enzymes, and ion channels) also known as pharmacodynamics genes as well as pharmacokinetic genes, which are genes involved in drug metabolism [11]. Despite significant efforts to gain a deeper understanding of all pharmacogenes, receptors PGx remains largely understudied. G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) are classical drug targets. GPCRs constitute the largest family of transmembrane proteins, transducing extracellular stimuli (hormones, neurotransmitters, and nutrients) into intracellular effects, thus controlling a plethora of bodily functions [12]. Although they serve as the most successful druggable protein family, as 35% of drugs target them, no GPCR variant is included in the drug labeling information [13]. Meanwhile, the genetic variation in a receptor tyrosine kinase, namely epidermal growth factor receptor (EGFR), whose overexpression was linked with cancer progression, is well defined in correlation with its inhibitors [13,14].
The integration of personalized medicine into common clinical practice has long been proposed, but challenges are yet to be defeated [15]. In this review, we will lay out the current status of PGx-informed pharmacotherapy regarding EGFR and emphasize the crucial significance of evaluating GPCR polymorphism in drug response.

2. Pharmacogenomics: Mechanism and Implementation

2.1. An Overview on Pharmacogenes Polymorphism

Pharmacogenomics (PGx) evaluates the influence of genetic variation on drug response. Although they are commonly used interchangeably, pharmacogenetics takes into account a single-gene effect, while PGx follows a more integrative approach by addressing the impact of the whole genome on drug treatment outcomes. Within the pharmacogenes, multiple types of genetic variations can arise due to single nucleotide polymorphisms (SNPs), indels (small insertions/deletions), inversions, duplications, or complex recombinations [16] (Figure 1). As a result, protein structure or function can be altered. The functional consequences of these mutations can result in an enhanced or novel protein activity in the case of gain-of-function (GoF) variants or in a diminished or non-existent activity owing to loss-of-function (LoF) variants [17]. Polymorphism in genes that encode drug-metabolizing enzymes or drug transport has a pharmacokinetic impact, while mutations in drug target genes (receptors, enzymes, and intracellular signaling proteins) result in an altered pharmacodynamics profile [18]. The most widely studied pharmacogenes are involved in drug metabolism, especially the well-known CYP450 family members, which are highly polymorphic, such as the CYP2D6 variants that have been thoroughly characterized [19]. On the contrary, pharmacodynamics drug–gene interactions are less understood [20]. Assessment of genetic variants that impact drug response relies on two approaches: the candidate gene approach (CGAS) or genome-wide association studies (GWAS). Both techniques explore the link between a phenotype (e.g., an adverse reaction or drug efficacy) and a genotype, although their study design is different.
The first one requires pre-specified genes known to be involved in drug pharmacology (drug metabolism or drug action). For example, warfarin dosing depends on the genetic polymorphism of two candidate genes: the metabolizing enzyme, CYP2C9, and the drug target, VKORC1 [24]. Since CGAS is a hypothesis-driven approach, it is more prone to bias if the wrong genes are chosen. Moreover, it is a self-limiting method because other important genes can be missed [25,26]. Meanwhile, GWAS employs a genome-wide screening strategy without the need for an a priori hypothesis. For instance, GWAS revealed that SLCO1B1 polymorphism predicts the risk of statin-induced myopathy, leading to improved adverse reaction management for patients who have been prescribed statins [27]. GWAS can identify novel mutated genes responsible for the observed phenotype, making it a useful approach for common variants, while CGAS is a method for studying rare variants [23]. Comparisons between these genetic associations are reviewed here [28], favoring GWAS. It should be noted that GWAS have their own limitations: (a) they can identify many genetic variants and can be cumbersome to determine the causal genes; (b) large sample sizes are mandatory for achieving statistical significance; and (c) population stratification can generate a false association [22,29,30]. Published GWAS are usually focused on understanding diseases rather than drug response. Hence, further PGx GWAS are vital for discovering relevant therapeutic biomarkers and the pathways involved in drug action [30].

2.2. Clinical Implementation of Pharmacogenomics

Implementation of PGx in clinical practice is a slow process, regardless of its proven role in drug therapy. Few hospitals or clinics from all over the world are applying the PGx tools into their practice, performing routine genetic testing for their patients [31,32,33,34,35]. The resistance of healthcare professionals against genotype-specific therapy arises from multiple reasons: (1) an apparent insufficient clinical utility; (2) scarcity of cost-effectiveness studies; (3) lack of standardized genotyping tests for each drug–gene pair; (4) difficulty in interpreting PGx tests; and (5) absence of a specific guideline or a step-by-step procedure for patients with a mutated pharmacogene [20]. The gold standard of evidence-based medicine is represented by randomized controlled trials (RCTs). In traditional medicine, prescribing decisions regarding commonly used drugs can rely on disease-oriented evidence implemented by case series rather than RCTs [36,37]. In addition, RCTs are costly, patient-specific factors (age, polypharmacy, and comorbidities) may not be taken into account, and rare variants can be overlooked [38]. Thus, in the case of PGx-guided therapy, confirmation of clinical utility cannot rely only on RCTs; however, alternative forms of evidence (retrospective studies and meta-analysis) may be sufficient if they are properly designed and analyzed. The overall cost for PGx-based treatment has decreased in the past few years due to technological advances, e.g., from single gene testing to a multigene panel [39]. PGx tests are not yet reimbursed due to a lack of widespread cost-effectiveness studies, although recent data demonstrate a positive economic outcome when PGx testing is performed [40,41]. Lastly, PGx implementation is stalled as a consequence of an absent harmonized guideline. A global network of experts (researchers and clinicians) and patients would help propel PGx research through translation into clinical practice [42]. Since 2005, the FDA has recommended the use of PGx data in drug development for new drug applications, reinforcing the inclusion of PGx information in drug labeling [13,43,44]. Labeling sections can include indications and usage, dosage and administration, adverse reactions, clinical pharmacology, clinical studies, warnings, and precautions, and even boxed warnings can be used to display PGx biomarkers [13]. Moreover, since 2020, the FDA has issued a Table of Pharmacogenetic Associations categorizing different gene–drug pairs with an emphasis on drug-metabolizing enzymes, drug transporters, and gene variants that can produce certain adverse reactions [6]. Likewise, the European Medicines Agency published its first PGx guideline in 2012 covering the impact of pharmacokinetic modifications due to genetic variation in key proteins [45]. Currently, there are several organizations whose purpose is to formulate recommendations to facilitate PGx application in clinical practice, such as the Clinical Pharmacogenetics Implementation Consortium (CPIC), the Dutch Pharmacogenetics Working Group (DPWG), the Canadian Pharmacogenomics Network for Drug Safety (CPNDS), and the French National Network (Réseau) of Pharmacogenetics (RNPGx) [46]. Each assembly has a distinctive profile and a different approach. Until now, CPIC emitted 26 guidelines providing guidance only for patients that already have PGx data, unlike DPWG, CPNDS, and RNPGx, which advise performing genotype testing in daily clinical practice, with RNPGx offering specific clinical characteristics for when it is suitable to test [46]. Considering this discrepancy, an important question is being raised: when and how to test? Ideally, genotype testing would be performed before prescribing, providing a patient-oriented, personalized guide to drug selection and dosage, reducing adverse reactions, and avoiding ineffective treatments [47]. This approach is called pre-emptive PGx and shows great potential for achieving genotype-based therapy [48,49,50,51,52,53,54]. Usually, a panel with different variants associated with commonly used drugs is tested and the information obtained is recorded electronically, generating a digital PGx profile for each patient serving as a starting point for any following medical problems. However, PGx tests are largely unregulated, unstandardized, and difficult to interpret. Based on high-quality and consistent evidence, PGx tests need to have the same gene and allele content linked with recommendations regarding medication selection and dosing [55].
Initiatives and efforts to harmonize PGx specialization worldwide have been made. The Association of Molecular Pathology (AMP), an international non-profit scientific society, provides information regarding the choice of alleles to be included for testing. The CPIC and DPWG are standardizing terms for PGx test results, in line with the Pharmacogene Variation Consortium (PharmVar), which also offers a universal PGx nomenclature. Pharmacogenomics Clinical Annotation Tool (PharmCAT) uses CPIC variants and assigns the corresponding allele for PGx test results interpretation, generating a PGx report. PharmGKB is an important resource for healthcare providers that aids in spreading information about genetic variants and their implications in drug response [56,57]. Moreover, globally, in the last 20 years, PGx education has been incorporated in medical and pharmacy schools, although some refinement is still needed (e.g., more hours of PGx education to be adopted, PGx as an independent pharmacy specialty, a more interdisciplinary perspective for better integrating PGx information, and creating E-learning programs for low-developed countries) [58].

3. The Influence of GPCR Genetic Variation on Drug Response

3.1. GPCR Pharmacogenetics Overview

GPCRs are the largest superfamily of membrane proteins in the human genome with over 800 members [59]. Currently, approximately 400 drugs approved by the FDA mediate their effect via 108 GPCRs. Moreover, novel GPCRs, which are not a target for any approved drug, are being investigated in clinical trials [60]. Although functionally diverse, the 7TM receptors (seven-transmembrane receptors) share a common design: an extracellular N-terminus, a C-terminal cytoplasmic domain, and seven transmembrane domains (TM1-7) joined together by three extracellular loops (ECL1-3) and three intracellular loops (ICL1-3) [59]. Upon ligand binding, a conformational shift takes place that favors the interaction with heterotrimeric G proteins. The Gα subunit has an intrinsic GTPase activity and transits between a GDP-bound inactive state and a GTP-bound active one. The GTP-bound Gα dissociates from the Gβγ subunit, triggering further cellular signaling. Based on the Gα subunit, G proteins are designated as Gαs, Gαi/o, Gαq/11, and Gα12/13 and regulate the activity of adenylyl cyclase, phospholipase C, or RhoGEFs [61]. Another GPCR interacting partner is represented by β-arrestins, which mediate desensitization and receptor internalization. Furthermore, β-arrestin-dependent, G protein-signaling has been described [62], but it is still under investigation regarding the G protein implication [63].
GPCR mutations were previously linked with disease occurrence, namely mutated rhodopsin receptor with retinitis pigmentosa [64], the calcium-sensing receptor with hypercalcemic syndromes [65], arginine vasopressin receptor 2 with nephrogenic diabetes insipidus [66], and many others [67,68,69,70]. Fewer studies exist regarding drug response. GPCR drug targets can be largely affected by missense mutations, followed by LoF variations that comprise nonsense mutations resulting in introducing a stop codon or frameshift mutations and copy number variations (deletions/duplications) [71]. The resulting functional implications can include decreased ligand binding, modified G protein selectivity or altered expression, localization, trafficking, or desensitization (Figure 2) [71].
The most targeted GPCRs, based on the number of approved drugs, are the adrenergic receptors, followed by the histamine receptors, serotonin, muscarinic, dopamine, and opioid receptors [60]. β-blockers, opioid analgesics, selective serotonin reuptake inhibitors, and second-generation antipsychotics that regulate dopaminergic and serotoninergic systems are among the most prescribed drugs in the US [75]. In the following subsections, the PGx of these receptors in correlation with drug reaction will be described and notable genetic variants are summarized in Table 1.

3.2. Antihypertension Therapy and β1-Adrenoreceptor Polymorphism

Adrenergic receptors are responsible for the regulation of the sympathetic nervous system, responding to endogenous catecholamines: adrenaline and noradrenaline. Ubiquitously expressed throughout the body, the nine receptor subtypes (α1A, α1B, α1D, α2A, α2B, α2C, β1, β2, and β3) have a multitude of indications as drug targets [97], but available PGx data focus mainly on β1-adrenergic blockade. Gene variants associated with dysregulated β1-adrenergic signaling are rs1801252:S49G (N-terminus) and rs1801253:R389G (helix 8) [98,99]. Greater reduction in blood pressure by metoprolol [76] or carvedilol [77] was observed for individuals with an R389R genotype than those who carried the R389G variant. Moreover, the S49R389/S49R389 diplotype can serve as a response predictor [76]. The ADRB1 Ser49-Arg389 haplotype was associated with an increased risk of all-cause death, especially in individuals treated with verapamil, contrary to atenolol-treated individuals, indicating a protective role of the β1-blocker [100]. However, atenolol produced a higher risk of adverse cardiovascular events in the case of individuals who have the ADRB1 S49G genotype [79].

3.3. Analgesics Treatment and µ1-Opioid Receptor Polymorphism

The µ1-opioid receptor (MOP) belongs to the class of opioid receptors that binds encephalin and β-endorphin-mediating analgesics effects. The classical µ1-opioid receptor agonist is morphine, but more potent synthetic ligands have been developed, such as fentanyl and derivatives serving as first-line therapy in moderate to severe pain management [101]. The most common SNP in the OPM1 gene is rs1799971:N40D, causing the loss of an N-glycosylation site [102], and being associated with a high sensitivity to opioids and increased susceptibility to opiate abuse [83,103].

3.4. Antipsychotic Drug Treatment and Dopamine/Serotonin Receptor Polymorphism

The D1 class of dopamine receptors includes D1 and D5 receptors, while the D2 class of dopamine receptors is comprised of D2, D3, and receptors. Antipsychotic drugs, regardless of their generation (typical—first generation; atypical—second generation) act primarily as D2 receptor antagonists. Intensive D2 receptor blockade, being linked with adverse events, is predominant in the first generation of antipsychotics. The second generation of antipsychotics also blocks the serotonin receptors (5HT1A, 5HT2A/2C, 5HT6, and 5HT7) [104]. We focused mainly on atypical antipsychotics that act as dopamine–serotonin antagonists (clozapine, risperidone, olanzapine, and aripiprazole).
DRD1 variants, rs4532 (A>48G) and rs5326 (G>94A), located in the 5′ UTR region are frequently studied [105]. Previously, the DRD1 rs4532 variant was associated with decreased clozapine response [106,107], but a meta-analysis showed a lack of correlation [108]. In the case of DRD2 polymorphisms, the most notable genetic variant is rs1799732: −141C Ins/Del, which is associated with reduced antipsychotic effects (Del allele compared with Ins/Ins genotype) [86]. Risperidone treatment response may be altered by genetic variants of DRD2, such as rs1801028:S311C, 141C Ins/Del (rs1799732), T939C (rs6275), rs6277, and TaqID [87]. While the rs6277 variant in DRD2 is associated with decreased response to ariprazole [109], the rs2514218 variant was correlated with better treatment response to aripiprazole or risperidone [110]. The rs6280:S9G variant is the most studied variant of the DRD3 gene, affecting the N-terminal extracellular domain [111]. This variant can serve as a predictor for olanzapine [112] or clozapine responses [113].
Atypical antipsychotic responses are similarly disturbed by mutated serotoninergic receptors. Risperidone efficacy was associated with rs6699866 in HTR6, while polymorphic HTR2C, HTR3D, and HTR5A have insignificant effects [114]. The HTR3B variant rs1176744:Y129S has an increased affinity for serotonin, with induction of dopamine signaling that opposes the antipsychotic effect, thus being associated with a decrease in treatment response [115]. During clozapine or olanzapine treatment, the HTR2A haplotype (rs6311:1438A, rs6312:783A, rs6313:102T, and rs6314:1354T) is correlated with a lower risk of developing metabolic abnormalities [116].

3.5. Antidepressants and Serotonin Receptors

Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed antidepressants, indicated for major depressive disorder and anxiety-related disorders. Inhibition of the serotonin transporter (SERT) in the presynaptic cleft, results in reduced serotonin uptake that can bind to 5HT1A autoreceptors [117,118]. Moreover, the SSRI vilazodone acts as a partial agonist for 5HT1A [119]. Other antidepressants that modulate the serotoninergic transmission are serotonin–noradrenaline reuptake inhibitors (SNRIs).
The HTR1A polymorphism in antidepressant response is still controversial; a few studies found no correlation [120,121], while others showed a better antidepressant response for patients expressing rs1364043, rs10042486, and rs6295 genetic variants when treated with milnacipran, fluvoxamine, fluoxetine, or paroxetine [91]. The rs6295 allele can also serve as a treatment predictor for citalopram response in older patients [122]. In the case of HTR2A, two common variants located in the promoter region have been widely studied, namely rs6311 and rs6313 [123]. These variants have been linked with higher treatment response; an increased risk of adverse reactions was associated with rs6311, while the rs6313 variant appears to have a protective effect [124]. A couple of studies reported no interdependency between the rs6311 and the antidepressant response [125,126,127], while for the rs6313 variant, a significant association was found [126].

4. EGFR, a PGx-Guided Therapy Model

EGFR, known as ErbB1/HER1, belongs to EGFR family together with ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4. We chose EGFR as an example for receptor PGx-informed therapy because (1) it is expressed in different cancer types, (2) its mutational landscape is well characterized, (3) aberrantly expressed EGFR is targeted by the majority of tyrosine kinase inhibitors (TKIs), and (4) for a drug target, it has a significant amount of PGx data [128,129,130,131].
EGFR signaling plays a pivotal role in regulating essential biological processes, including cell proliferation, differentiation, migration, adhesion, and survival [132]. As a membrane-spanning receptor, EGFR connects extracellular stimuli to the intracellular signaling pathways. Structurally, EGFR features four extracellular domains (I, II, III, and IV), one transmembrane domain, one juxtamembrane domain, and one tyrosine kinase domain, followed by a flexible regulatory C-terminus (Figure 3) [133]. Under resting conditions, EGFR exists as a monomer. Upon ligand binding to the extracellular domain, dimerization of the receptor occurs, followed by autophosphorylation and initiation of a downstream signaling cascade, namely the rat sarcoma (Ras)/mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB) pathways [132].
Renowned as an oncogene, the aberrantly expressed EGFR represents a malignancy hallmark, being more prevalent in lung, colorectal, and brain cancers [136]. Usually, EGFR oncogenicity is characterized by overexpression, gene amplification, or activating mutations [137]. The majority of the mutations occur within the EGFR exons 18 to 21, which encode a portion of the EGFR kinase domain (Figure 3) [138,139,140]. The purpose of EGFR-targeted therapy is to detect EGFR-positive cancers and existent mutations and to combat frequently acquired drug resistance. Deviant EGFR is managed by two treatment directions, tyrosine kinase inhibitors (TKIs) and monoclonal antibodies (Mabs) (as described in Table 2, which contains information from the FDA Table of Pharmacogenomic Biomarkers in Drug Labeling). Among the approved EGFR-TKIs, gefitinib and erlotinib belong to the first generation of EGFR inhibitors. They work by binding reversibly to the TK domain of the EGFR, blocking ATP binding, thereby halting EGFR activation and cellular proliferation. Afatinib and dacomitinib are members of the second generation of EGFR-TKIs, binding irreversibly to the EGFR, thus inhibiting its kinase activity, while osimertinib binds covalently to the cysteine residue in the EGFR [141,142]. Mobocertinib is designed to target EGFR exon 20 insertions [143]. Monoclonal antibodies (cetuximab, panitumumab, and amivantamab) bind to the extracellular domain III of EGFR, promoting ligand blocking, receptor–antibody complex internalization, and degradation [144]. Of note, kinase-activating mutations cannot disturb the activity of these antibodies since their binding site is in the extracellular domain.
According to PharmGKB, the EGFR is a Very Important Pharmacogene (VIP). Currently, on the PharmGKB database, there are 21 clinical annotations regarding EGFR, 13 of them involving the interaction between EGFR inhibitors and their target (Table 3) [8,9].
The FDA Table of Pharmacogenomic Biomarkers in Drug Labeling offers a perspective on available EGFR-targeted drugs and their PGx information, being indication-oriented (using an FDA-approved test to identify the specific drug targeted mutation for treatment). The clinical annotations from PharmGKB resemble the FDA Table of Pharmacogenetic Associations, offering information about the interaction between a genetic variant and a drug. Each pair contains the “Details” section where a specific allele is linked with an increased or decreased response, adverse reaction, or even with the likelihood of acquired resistance. Furthermore, the level of evidence is provided, with level 3 being the most assigned. In contrast, the FDA Table of Pharmacogenetic Associations contains gene–drug interactions that impact drug metabolism and have sufficient scientific evidence. At the moment, EGFR is not included in this table. Therefore, healthcare providers should take into account all available resources to guide and treat each patient individually based on their genetic makeup and to monitor therapy outcomes.
The major indication of EGFR-targeted therapy is represented by non-small-cell lung cancer (NSCLC). In NSCLC, the effectiveness of conventional chemotherapy treatments does not exceed 40%, being also associated with a high degree of toxicity and poor prognosis. Abnormal EGFR signaling contributes to the oncogenic phenotype in over half of NSCLC patients. TKIs interact with the aberrantly expressed receptor, significantly increasing the survival rate of patients and exceeding classical chemotherapy response levels in NSCLC patients [146]. Of all EGFR mutations, 45% are deletions or insertions (amino-acid residues 747 to 752) of exon 19, with the most common being delE746_A750. Additionally, the exon 21 point mutation L858R accounts for roughly 40% of EGFR mutations. The third most common type (10% of all EGFR mutations) consists of in-frame insertions and indels in exon 20 [147]. Exons 18, 19, and 21 mutations are sensitivity predictors of EGFR-TKI therapy, while mutations in exon 20 are generally resistant [148]. The emergence of EGFR T790M and C797S mutations (exon 20) has led to rapid resistance development against first-, second-, and third-generation EGFR-TKIs. The C797S mutation specifically hinders irreversible EGFR inhibitors’ ability to bind covalently to the kinase. Additionally, the rise of EGFR double and triple mutations poses challenges to the therapeutic efficacy of EGFR-TKIs, emphasizing the continuous necessity for potent new inhibitors [141].
While promising results are limited for other types of cancer besides NSCLC, erlotinib in combination with gemcitabine received FDA approval for use in the treatment of locally advanced, unresectable, or metastatic pancreatic cancer on 2 November 2005. In pancreatic cancer, EGFR overexpression is correlated with advanced disease, poor survival, and metastasis. Combining EGFR inhibitors with chemotherapy results in the inhibition of tumor-induced angiogenesis, promotion of tumor cell apoptosis, and tumor regression in xenograft models [149]. The non-frameshift deletion in exon 19 and the L858R point mutation in exon 21 are known as EGFR-sensitive mutations and respond well to EGFR-TKIs [150]. However, these mutations are rare in pancreatic ductal adenocarcinoma (PDAC) [151]. Similarly, in colorectal cancer, the main contributor to poor prognosis is EGFR overexpression [152]. Hence, blocking EGFR signaling is the ideal approach. Cetuximab and panitumumab are both indicated for EGFR-expressing, RAS wild-type metastatic colorectal cancer, serving as monotherapy or in combination with chemotherapy [153]. Additionally, cetuximab is also indicated for head and neck cancer in combination with radiation therapy, antineoplastic agents, or alone for recurrent disease or metastasis [154]. However, in this case, cetuximab therapy has a poor response rate, regardless of high EGFR gene amplification. Less than 5% of head and neck cancers present EGFR mutations, making this cancer type particularly difficult to treat [155]. Mutations or genetic variations in EGFR alone cannot account for all the differences in cancer patients’ responses to EGFR-targeted treatments. Beyond the EGFR genotype, genetic variances in other components of the signaling pathway downstream of EGFR or in other receptor tyrosine kinase pathways can significantly affect the effectiveness of specific EGFR-targeted therapies [156,157,158], emphasizing the personalized medicine approach of treating individual patients based on their particular physiology and needs.

5. Conclusions

In the digital era, personalized medicine can be achieved in order to provide patients with tailored therapies. By employing PGx tools, each patient can be treated based on their unique genetic makeup, reducing the risk of adverse reactions or treatment failure. However, it is still unclear when the PGx tests are recommended and how to apply and interpret them. Despite great efforts, the implementation of PGx-informed therapy is still incomplete. Moreover, the healthcare authorities (FDA and EMA) issued guidelines regarding only pharmacokinetic genetic variants due to a lack of sufficient evidence. One problem may be the inconsistency noticed between different studies that analyzed the same gene–drug pair. Thus, standardization is also an absolute requirement to further advance the field of PGx. In the case of commonly prescribed drugs, genetic variants that affect drug response are still understudied. Here, we used an RTK EGFR as a prototype for emphasizing the crucial role of determining receptors’ genetic influence on drug response. As more than one-third of approved drugs target GPCRs, a complete mutational landscape must be generated. We focused only on genetic variants that affect targets, without including studies evaluating the downstream signaling partners. Cellular signaling functions as an interconnected machinery. Hence, the net drug response may be a consequence of different altered proteins from the same pathway or from intersected pathways.

Author Contributions

Conceptualization, S.A.A., C.-E.D.-P. and L.P.; methodology, C.-E.D.-P., S.A.A., M.-A.C. and A.M.V.; formal analysis, S.A.A., C.-E.D.-P., M.-A.C. and A.M.V.; investigation, S.A.A., C.-E.D.-P., M.-A.C. and A.M.V.; resources, C.-E.D.-P. and L.P.; writing—original draft preparation, S.A.A., C.-E.D.-P., M.-A.C. and A.M.V.; writing—review and editing, L.P., M.V.G., C.-E.D.-P. and V.A.; visualization, S.A.A., C.-E.D.-P., L.P., M.-A.C. and A.M.V.; supervision, L.P., M.V.G., C.-E.D.-P. and V.A.; project administration, L.P., M.V.G., C.-E.D.-P. and V.A.; funding acquisition, C.-E.D.-P. and V.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded through the Institutional Program Publish not Perish, 2024, and by the Institutional Development Fund, CNFIS-FDI-2024-F-0570.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

This scientific paper was financially supported by the “Carol Davila” University of Medicine and Pharmacy Bucharest through Contract No. CNFIS-FDI-2024-F-0570.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. International Human Genome Sequencing Consortium, Finishing the euchromatic sequence of the human genome. Nature 2004, 431, 931–945. [CrossRef]
  2. Lu, C.; Ahmed, R.; Lamri, A.; Anand, S.S. Use of race, ethnicity, and ancestry data in health research. PLoS Glob. Public Health 2022, 2, e0001060. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, R.C.; Wang, Z. Precision Medicine: Disease Subtyping and Tailored Treatment. Cancers 2023, 15, 3837. [Google Scholar] [CrossRef] [PubMed]
  4. Hulsen, T.; Jamuar, S.S.; Moody, A.R.; Karnes, J.H.; Varga, O.; Hedensted, S.; Spreafico, R.; Hafler, D.A.; McKinney, E.F. From Big Data to Precision Medicine. Front. Med. 2019, 6, 34. [Google Scholar] [CrossRef]
  5. White, C.; Scott, R.; Paul, C.L.; Ackland, S.P. Pharmacogenomics in the era of personalised medicine. Med. J. Aust. 2022, 217, 510–513. [Google Scholar] [CrossRef] [PubMed]
  6. US Food and Drug Administration. Table of Pharmacogenetic Associations. Available online: https://www.fda.gov/medical-devices/precision-medicine/table-pharmacogenetic-associations (accessed on 28 March 2024).
  7. Klein, T.E.; Chang, J.T.; Cho, M.K.; Easton, K.L.; Fergerson, R.; Hewett, M.; Lin, Z.; Liu, Y.; Liu, S.; Oliver, D.E.; et al. Integrating genotype and phenotype information: An overview of the PharmGKB project. Pharmacogenetics Research Network and Knowledge Base. Pharmacogenom. J. 2001, 1, 167–170. [Google Scholar] [CrossRef]
  8. Whirl-Carrillo, M.; Huddart, R.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Whaley, R.; Klein, T.E. An Evidence-Based Framework for Evaluating Pharmacogenomics Knowledge for Personalized Medicine. Clin. Pharmacol. Ther. 2021, 110, 563–572. [Google Scholar] [CrossRef]
  9. Whirl-Carrillo, M.; McDonagh, E.M.; Hebert, J.M.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Altman, R.B.; Klein, T.E. Pharmacogenomics knowledge for personalized medicine. Clin. Pharmacol. Ther. 2012, 92, 414–417. [Google Scholar] [CrossRef]
  10. Pritchard, D.; Patel, J.N.; Stephens, L.E.; McLeod, H.L. Comparison of FDA Table of Pharmacogenetic Associations and Clinical Pharmacogenetics Implementation Consortium guidelines. Am. J. Health Syst. Pharm. 2022, 79, 993–1005. [Google Scholar] [CrossRef]
  11. Rukov, J.L.; Vinther, J.; Shomron, N. Pharmacogenomics genes show varying perceptibility to microRNA regulation. Pharmacogenet. Genom. 2011, 21, 251–262. [Google Scholar] [CrossRef]
  12. Rosenbaum, D.M.; Rasmussen, S.G.; Kobilka, B.K. The structure and function of G-protein-coupled receptors. Nature 2009, 459, 356–363. [Google Scholar] [CrossRef] [PubMed]
  13. US Food and Drug Administration. Table of Pharmacogenomic Biomarkers in Drug Labeling. Available online: https://www.fda.gov/drugs/science-and-research-drugs/table-pharmacogenomic-biomarkers-drug-labeling (accessed on 28 March 2024).
  14. Jimeno, A.; Hidalgo, M. Pharmacogenomics of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors. Biochim. Biophys. Acta 2006, 1766, 217–229. [Google Scholar] [CrossRef] [PubMed]
  15. Abou Diwan, E.; Zeitoun, R.I.; Abou Haidar, L.; Cascorbi, I.; Khoueiry Zgheib, N. Implementation and obstacles of pharmacogenetics in clinical practice: An international survey. Br. J. Clin. Pharmacol. 2019, 85, 2076–2088. [Google Scholar] [CrossRef]
  16. He, Y.; Hoskins, J.M.; McLeod, H.L. Copy number variants in pharmacogenetic genes. Trends Mol. Med. 2011, 17, 244–251. [Google Scholar] [CrossRef] [PubMed]
  17. Stein, D.; Kars, M.E.; Wu, Y.; Bayrak, C.S.; Stenson, P.D.; Cooper, D.N.; Schlessinger, A.; Itan, Y. Genome-wide prediction of pathogenic gain- and loss-of-function variants from ensemble learning of a diverse feature set. Genome Med. 2023, 15, 103. [Google Scholar] [CrossRef] [PubMed]
  18. Roden, D.M.; McLeod, H.L.; Relling, M.V.; Williams, M.S.; Mensah, G.A.; Peterson, J.F.; Van Driest, S.L. Pharmacogenomics. Lancet 2019, 394, 521–532. [Google Scholar] [CrossRef] [PubMed]
  19. Zhou, Y.; Lauschke, V.M. The genetic landscape of major drug metabolizing cytochrome P450 genes-an updated analysis of population-scale sequencing data. Pharmacogenom. J. 2022, 22, 284–293. [Google Scholar] [CrossRef]
  20. Pirmohamed, M. Pharmacogenomics: Current status and future perspectives. Nat. Rev. Genet. 2023, 24, 350–362. [Google Scholar] [CrossRef]
  21. Ramirez-Bello, J.; Jimenez-Morales, M. Functional implications of single nucleotide polymorphisms (SNPs) in protein-coding and non-coding RNA genes in multifactorial diseases. Gac. Med. Mex. 2017, 153, 238–250. [Google Scholar]
  22. Tam, V.; Patel, N.; Turcotte, M.; Bosse, Y.; Pare, G.; Meyre, D. Benefits and limitations of genome-wide association studies. Nat. Rev. Genet. 2019, 20, 467–484. [Google Scholar] [CrossRef]
  23. David, S. A current guide to candidate gene association studies. Trends Genet. 2021, 37, 1056–1059. [Google Scholar] [CrossRef] [PubMed]
  24. Hertz, D.L.; Arwood, M.J.; Stocco, G.; Singh, S.; Karnes, J.H.; Ramsey, L.B. Planning and Conducting a Pharmacogenetics Association Study. Clin. Pharmacol. Ther. 2021, 110, 688–701. [Google Scholar] [CrossRef] [PubMed]
  25. Chan, H.T.; Chin, Y.M.; Low, S.K. The Roles of Common Variation and Somatic Mutation in Cancer Pharmacogenomics. Oncol. Ther. 2019, 7, 1–32. [Google Scholar] [CrossRef]
  26. Duarte, R.R.R.; Brentani, H.; Powell, T.R. Ditching candidate gene association studies: Lessons from psychiatric genetics. Braz. J. Psychiatry 2021, 43, 342–344. [Google Scholar] [CrossRef] [PubMed]
  27. Group, S.C.; Link, E.; Parish, S.; Armitage, J.; Bowman, L.; Heath, S.; Matsuda, F.; Gut, I.; Lathrop, M.; Collins, R. SLCO1B1 variants and statin-induced myopathy—A genomewide study. N. Engl. J. Med. 2008, 359, 789–799. [Google Scholar] [CrossRef]
  28. Linskey, D.W.; Linskey, D.C.; McLeod, H.L.; Luzum, J.A. The need to shift pharmacogenetic research from candidate gene to genome-wide association studies. Pharmacogenomics 2021, 22, 1143–1150. [Google Scholar] [CrossRef] [PubMed]
  29. Qi, T.; Song, L.; Guo, Y.; Chen, C.; Yang, J. From genetic associations to genes: Methods, applications, and challenges. Trends Genet. 2024, 40, 642–667. [Google Scholar] [CrossRef]
  30. Giacomini, K.M.; Yee, S.W.; Mushiroda, T.; Weinshilboum, R.M.; Ratain, M.J.; Kubo, M. Genome-wide association studies of drug response and toxicity: An opportunity for genome medicine. Nat. Rev. Drug Discov. 2017, 16, 70. [Google Scholar] [CrossRef]
  31. Shuldiner, A.R.; Relling, M.V.; Peterson, J.F.; Hicks, J.K.; Freimuth, R.R.; Sadee, W.; Pereira, N.L.; Roden, D.M.; Johnson, J.A.; Klein, T.E.; et al. The Pharmacogenomics Research Network Translational Pharmacogenetics Program: Overcoming challenges of real-world implementation. Clin. Pharmacol. Ther. 2013, 94, 207–210. [Google Scholar] [CrossRef] [PubMed]
  32. Cavallari, L.H.; Lee, C.R.; Duarte, J.D.; Nutescu, E.A.; Weitzel, K.W.; Stouffer, G.A.; Johnson, J.A. Implementation of inpatient models of pharmacogenetics programs. Am. J. Health Syst. Pharm. 2016, 73, 1944–1954. [Google Scholar] [CrossRef]
  33. van der Wouden, C.H.; Cambon-Thomsen, A.; Cecchin, E.; Cheung, K.C.; Davila-Fajardo, C.L.; Deneer, V.H.; Dolzan, V.; Ingelman-Sundberg, M.; Jonsson, S.; Karlsson, M.O.; et al. Implementing Pharmacogenomics in Europe: Design and Implementation Strategy of the Ubiquitous Pharmacogenomics Consortium. Clin. Pharmacol. Ther. 2017, 101, 341–358. [Google Scholar] [CrossRef] [PubMed]
  34. Borobia, A.M.; Dapia, I.; Tong, H.Y.; Arias, P.; Munoz, M.; Tenorio, J.; Hernandez, R.; Garcia Garcia, I.; Gordo, G.; Ramirez, E.; et al. Clinical Implementation of Pharmacogenetic Testing in a Hospital of the Spanish National Health System: Strategy and Experience over 3 Years. Clin. Transl. Sci. 2018, 11, 189–199. [Google Scholar] [CrossRef] [PubMed]
  35. Wick, J.A.; Schmidlen, T.; Grande, K.; Moretz, C.; Ashcraft, K.; Green, J.; Moyer, N.; Blaxall, B.C. Implementing comprehensive pharmacogenomics in a community hospital-associated primary care setting. J. Am. Pharm. Assoc. 2023, 63, 188–192. [Google Scholar] [CrossRef] [PubMed]
  36. Munar, M.Y.; Singh, H. Drug dosing adjustments in patients with chronic kidney disease. Am. Fam. Phys. 2007, 75, 1487–1496. [Google Scholar]
  37. Gloy, V.; Schmitt, A.M.; Dublin, P.; Hirt, J.; Axfors, C.; Kuk, H.; Pereira, T.V.; Locher, C.; Caquelin, L.; Walter-Claudi, M.; et al. The evidence base of US Food and Drug Administration approvals of novel cancer therapies from 2000 to 2020. Int. J. Cancer 2023, 152, 2474–2484. [Google Scholar] [CrossRef] [PubMed]
  38. Huddart, R.; Sangkuhl, K.; Whirl-Carrillo, M.; Klein, T.E. Are Randomized Controlled Trials Necessary to Establish the Value of Implementing Pharmacogenomics in the Clinic? Clin. Pharmacol. Ther. 2019, 106, 284–286. [Google Scholar] [CrossRef]
  39. Marrero, R.J.; Cicali, E.J.; Arwood, M.J.; Eddy, E.; DeRemer, D.; Ramnaraign, B.H.; Daily, K.C.; Jones, D., Jr.; Cook, K.J.; Cavallari, L.H.; et al. How to Transition from Single-Gene Pharmacogenetic Testing to Preemptive Panel-Based Testing: A Tutorial. Clin. Pharmacol. Ther. 2020, 108, 557–565. [Google Scholar] [CrossRef]
  40. Morris, S.A.; Alsaidi, A.T.; Verbyla, A.; Cruz, A.; Macfarlane, C.; Bauer, J.; Patel, J.N. Cost Effectiveness of Pharmacogenetic Testing for Drugs with Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines: A Systematic Review. Clin. Pharmacol. Ther. 2022, 112, 1318–1328. [Google Scholar] [CrossRef]
  41. van der Wouden, C.H.; Marck, H.; Guchelaar, H.J.; Swen, J.J.; van den Hout, W.B. Cost-Effectiveness of Pharmacogenomics-Guided Prescribing to Prevent Gene-Drug-Related Deaths: A Decision-Analytic Model. Front. Pharmacol. 2022, 13, 918493. [Google Scholar] [CrossRef]
  42. Chenoweth, M.J.; Giacomini, K.M.; Pirmohamed, M.; Hill, S.L.; van Schaik, R.H.N.; Schwab, M.; Shuldiner, A.R.; Relling, M.V.; Tyndale, R.F. Global Pharmacogenomics Within Precision Medicine: Challenges and Opportunities. Clin. Pharmacol. Ther. 2020, 107, 57–61. [Google Scholar] [CrossRef]
  43. US Food and Drug Administration Guidance for Industry. Pharmacogenomic Data Submissions. Available online: https://www.fda.gov/media/72420/download (accessed on 9 June 2024).
  44. US Food and Drug Administration Guidance for Industry. Clinical Pharmacogenomics: Premarket Evaluation in Early-Phase Clinical Studies and Recommendations for Labeling. Available online: https://www.fda.gov/media/84923/download (accessed on 9 June 2024).
  45. EMA: Guideline on the Use of Pharmacogenetic Methodologies in the Pharmacokinetic Evaluation of Medicinal Products. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-use-pharmacogenetic-methodologies-pharmacokinetic-evaluation-medicinal-products_en.pdf (accessed on 9 June 2024).
  46. Abdullah-Koolmees, H.; van Keulen, A.M.; Nijenhuis, M.; Deneer, V.H.M. Pharmacogenetics Guidelines: Overview and Comparison of the DPWG, CPIC, CPNDS, and RNPGx Guidelines. Front. Pharmacol. 2020, 11, 595219. [Google Scholar] [CrossRef]
  47. Chenchula, S.; Atal, S.; Uppugunduri, C.R.S. A review of real-world evidence on preemptive pharmacogenomic testing for preventing adverse drug reactions: A reality for future health care. Pharmacogenom. J. 2024, 24, 9. [Google Scholar] [CrossRef]
  48. O’Donnell, P.H.; Bush, A.; Spitz, J.; Danahey, K.; Saner, D.; Das, S.; Cox, N.J.; Ratain, M.J. The 1200 patients project: Creating a new medical model system for clinical implementation of pharmacogenomics. Clin. Pharmacol. Ther. 2012, 92, 446–449. [Google Scholar] [CrossRef] [PubMed]
  49. Gottesman, O.; Kuivaniemi, H.; Tromp, G.; Faucett, W.A.; Li, R.; Manolio, T.A.; Sanderson, S.C.; Kannry, J.; Zinberg, R.; Basford, M.A.; et al. The Electronic Medical Records and Genomics (eMERGE) Network: Past, present, and future. Genet. Med. 2013, 15, 761–771. [Google Scholar] [CrossRef]
  50. Gottesman, O.; Scott, S.A.; Ellis, S.B.; Overby, C.L.; Ludtke, A.; Hulot, J.S.; Hall, J.; Chatani, K.; Myers, K.; Kannry, J.L.; et al. The CLIPMERGE PGx Program: Clinical implementation of personalized medicine through electronic health records and genomics-pharmacogenomics. Clin. Pharmacol. Ther. 2013, 94, 214–217. [Google Scholar] [CrossRef] [PubMed]
  51. Hoffman, J.M.; Haidar, C.E.; Wilkinson, M.R.; Crews, K.R.; Baker, D.K.; Kornegay, N.M.; Yang, W.; Pui, C.H.; Reiss, U.M.; Gaur, A.H.; et al. PG4KDS: A model for the clinical implementation of pre-emptive pharmacogenetics. Am. J. Med. Genet. Part C Semin. Med. Genet. 2014, 166C, 45–55. [Google Scholar] [CrossRef] [PubMed]
  52. Norris, M.; Dalton, R.; Alam, B.; Eddy, E.; Nguyen, K.A.; Cavallari, L.H.; Sumfest, J.; Wiisanen, K.; Cicali, E.J. Lessons from clinical implementation of a preemptive pharmacogenetic panel as part of a testing pilot program with an employer-sponsored medical plan. Front. Genet. 2023, 14, 1249003. [Google Scholar] [CrossRef]
  53. Luczak, T.S.; Schillo, P.J.; Renier, C.M.; Waring, S.C.; Friday, B.B. Feasibility of preemptive pharmacogenetic testing in colorectal cancer patients within a community oncology setting. J. Oncol. Pharm. Pract. 2022, 28, 842–849. [Google Scholar] [CrossRef]
  54. Lteif, C.; Eddy, E.; Terrell, J.; Cavallari, L.H.; Malaty, J.; Duarte, J.D. Feasibility of preemptive pharmacogenetic testing and improvement of medication treatment satisfaction among medically underserved patients. Clin. Transl. Sci. 2024, 17, e13692. [Google Scholar] [CrossRef]
  55. Maruf, A.A.; Bousman, C.A. Approaches and hurdles of implementing pharmacogenetic testing in the psychiatric clinic. PCN Rep. 2022, 1, e26. [Google Scholar] [CrossRef]
  56. Kabbani, D.; Akika, R.; Wahid, A.; Daly, A.K.; Cascorbi, I.; Zgheib, N.K. Pharmacogenomics in practice: A review and implementation guide. Front. Pharmacol. 2023, 14, 1189976. [Google Scholar] [CrossRef] [PubMed]
  57. Luzum, J.A.; Petry, N.; Taylor, A.K.; Van Driest, S.L.; Dunnenberger, H.M.; Cavallari, L.H. Moving Pharmacogenetics Into Practice: It’s All About the Evidence! Clin. Pharmacol. Ther. 2021, 110, 649–661. [Google Scholar] [CrossRef] [PubMed]
  58. Karas Kuzelicki, N.; Prodan Zitnik, I.; Gurwitz, D.; Llerena, A.; Cascorbi, I.; Siest, S.; Simmaco, M.; Ansari, M.; Pazzagli, M.; Di Resta, C.; et al. Pharmacogenomics education in medical and pharmacy schools: Conclusions of a global survey. Pharmacogenomics 2019, 20, 643–657. [Google Scholar] [CrossRef]
  59. Zhang, M.; Chen, T.; Lu, X.; Lan, X.; Chen, Z.; Lu, S. G protein-coupled receptors (GPCRs): Advances in structures, mechanisms, and drug discovery. Signal Transduct. Target. Ther. 2024, 9, 88. [Google Scholar] [CrossRef]
  60. Hauser, A.S.; Attwood, M.M.; Rask-Andersen, M.; Schioth, H.B.; Gloriam, D.E. Trends in GPCR drug discovery: New agents, targets and indications. Nat. Rev. Drug Discov. 2017, 16, 829–842. [Google Scholar] [CrossRef] [PubMed]
  61. Hilger, D.; Masureel, M.; Kobilka, B.K. Structure and dynamics of GPCR signaling complexes. Nat. Struct. Mol. Biol. 2018, 25, 4–12. [Google Scholar] [CrossRef]
  62. Jean-Charles, P.Y.; Kaur, S.; Shenoy, S.K. G Protein-Coupled Receptor Signaling Through beta-Arrestin-Dependent Mechanisms. J. Cardiovasc. Pharmacol. 2017, 70, 142–158. [Google Scholar] [CrossRef] [PubMed]
  63. Grundmann, M.; Merten, N.; Malfacini, D.; Inoue, A.; Preis, P.; Simon, K.; Ruttiger, N.; Ziegler, N.; Benkel, T.; Schmitt, N.K.; et al. Lack of beta-arrestin signaling in the absence of active G proteins. Nat. Commun. 2018, 9, 341. [Google Scholar] [CrossRef]
  64. Dryja, T.P.; McGee, T.L.; Reichel, E.; Hahn, L.B.; Cowley, G.S.; Yandell, D.W.; Sandberg, M.A.; Berson, E.L. A point mutation of the rhodopsin gene in one form of retinitis pigmentosa. Nature 1990, 343, 364–366. [Google Scholar] [CrossRef] [PubMed]
  65. Guarnieri, V.; Canaff, L.; Yun, F.H.; Scillitani, A.; Battista, C.; Muscarella, L.A.; Wong, B.Y.; Notarangelo, A.; D’Agruma, L.; Sacco, M.; et al. Calcium-sensing receptor (CASR) mutations in hypercalcemic states: Studies from a single endocrine clinic over three years. J. Clin. Endocrinol. Metab. 2010, 95, 1819–1829. [Google Scholar] [CrossRef]
  66. Spanakis, E.; Milord, E.; Gragnoli, C. AVPR2 variants and mutations in nephrogenic diabetes insipidus: Review and missense mutation significance. J. Cell. Physiol. 2008, 217, 605–617. [Google Scholar] [CrossRef] [PubMed]
  67. Luo, J.; Sun, P.; Siwko, S.; Liu, M.; Xiao, J. The role of GPCRs in bone diseases and dysfunctions. Bone Res. 2019, 7, 19. [Google Scholar] [CrossRef]
  68. Liu, T.; Ji, R.L.; Tao, Y.X. Naturally occurring mutations in G protein-coupled receptors associated with obesity and type 2 diabetes mellitus. Pharmacol. Ther. 2022, 234, 108044. [Google Scholar] [CrossRef]
  69. Fukami, M.; Suzuki, E.; Igarashi, M.; Miyado, M.; Ogata, T. Gain-of-function mutations in G-protein-coupled receptor genes associated with human endocrine disorders. Clin. Endocrinol. 2018, 88, 351–359. [Google Scholar] [CrossRef]
  70. Yang, L.K.; Hou, Z.S.; Tao, Y.X. Biased signaling in naturally occurring mutations of G protein-coupled receptors associated with diverse human diseases. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 165973. [Google Scholar] [CrossRef]
  71. Hauser, A.S.; Chavali, S.; Masuho, I.; Jahn, L.J.; Martemyanov, K.A.; Gloriam, D.E.; Babu, M.M. Pharmacogenomics of GPCR Drug Targets. Cell 2018, 172, 41–54.e19. [Google Scholar] [CrossRef]
  72. Basith, S.; Cui, M.; Macalino, S.J.Y.; Park, J.; Clavio, N.A.B.; Kang, S.; Choi, S. Exploring G Protein-Coupled Receptors (GPCRs) Ligand Space via Cheminformatics Approaches: Impact on Rational Drug Design. Front. Pharmacol. 2018, 9, 128. [Google Scholar] [CrossRef]
  73. Edward Zhou, X.; Melcher, K.; Eric Xu, H. Structural biology of G protein-coupled receptor signaling complexes. Protein Sci. 2019, 28, 487–501. [Google Scholar] [CrossRef]
  74. Schoneberg, T.; Liebscher, I. Mutations in G Protein-Coupled Receptors: Mechanisms, Pathophysiology and Potential Therapeutic Approaches. Pharmacol. Rev. 2021, 73, 89–119. [Google Scholar] [CrossRef]
  75. Fuentes, A.V.; Pineda, M.D.; Venkata, K.C.N. Comprehension of Top 200 Prescribed Drugs in the US as a Resource for Pharmacy Teaching, Training and Practice. Pharmacy 2018, 6, 43. [Google Scholar] [CrossRef]
  76. Johnson, J.A.; Zineh, I.; Puckett, B.J.; McGorray, S.P.; Yarandi, H.N.; Pauly, D.F. Beta 1-adrenergic receptor polymorphisms and antihypertensive response to metoprolol. Clin. Pharmacol. Ther. 2003, 74, 44–52. [Google Scholar] [CrossRef]
  77. Si, D.; Wang, J.; Xu, Y.; Chen, X.; Zhang, M.; Zhou, H. Association of common polymorphisms in beta1-adrenergic receptor with antihypertensive response to carvedilol. J. Cardiovasc. Pharmacol. 2014, 64, 306–309. [Google Scholar] [CrossRef]
  78. Fayed, M.S.; Saleh, M.A.; Sabri, N.A.; Elkholy, A.A. beta1-adrenergic receptor polymorphisms: A possible genetic predictor of bisoprolol response in acute coronary syndrome. Future Sci. OA 2023, 9, FSO895. [Google Scholar] [CrossRef]
  79. Magvanjav, O.; McDonough, C.W.; Gong, Y.; McClure, L.A.; Talbert, R.L.; Horenstein, R.B.; Shuldiner, A.R.; Benavente, O.R.; Mitchell, B.D.; Johnson, J.A.; et al. Pharmacogenetic Associations of beta1-Adrenergic Receptor Polymorphisms With Cardiovascular Outcomes in the SPS3 Trial (Secondary Prevention of Small Subcortical Strokes). Stroke 2017, 48, 1337–1343. [Google Scholar] [CrossRef]
  80. Turner, S.; Francis, B.; Vijverberg, S.; Pino-Yanes, M.; Maitland-van der Zee, A.H.; Basu, K.; Bignell, L.; Mukhopadhyay, S.; Tavendale, R.; Palmer, C.; et al. Childhood asthma exacerbations and the Arg16 beta2-receptor polymorphism: A meta-analysis stratified by treatment. J. Allergy Clin. Immunol. 2016, 138, 107–113.e105. [Google Scholar] [CrossRef] [PubMed]
  81. Bandaru, S.; Alvala, M.; Nayarisseri, A.; Sharda, S.; Goud, H.; Mundluru, H.P.; Singh, S.K. Molecular dynamic simulations reveal suboptimal binding of salbutamol in T164I variant of beta2 adrenergic receptor. PLoS ONE 2017, 12, e0186666. [Google Scholar] [CrossRef]
  82. Yang, Y.Y.; Lin, H.C.; Lee, W.P.; Chu, C.J.; Lin, M.W.; Lee, F.Y.; Hou, M.C.; Jap, J.S.; Lee, S.D. Association of the G-protein and alpha2-adrenergic receptor gene and plasma norepinephrine level with clonidine improvement of the effects of diuretics in patients with cirrhosis with refractory ascites: A randomised clinical trial. Gut 2010, 59, 1545–1553. [Google Scholar] [CrossRef]
  83. Bond, C.; LaForge, K.S.; Tian, M.; Melia, D.; Zhang, S.; Borg, L.; Gong, J.; Schluger, J.; Strong, J.A.; Leal, S.M.; et al. Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: Possible implications for opiate addiction. Proc. Natl. Acad. Sci. USA 1998, 95, 9608–9613. [Google Scholar] [CrossRef]
  84. Mura, E.; Govoni, S.; Racchi, M.; Carossa, V.; Ranzani, G.N.; Allegri, M.; van Schaik, R.H. Consequences of the 118A>G polymorphism in the OPRM1 gene: Translation from bench to bedside? J. Pain Res. 2013, 6, 331–353. [Google Scholar] [CrossRef]
  85. Knapman, A.; Santiago, M.; Connor, M. Buprenorphine signalling is compromised at the N40D polymorphism of the human mu opioid receptor in vitro. Br. J. Pharmacol. 2014, 171, 4273–4288. [Google Scholar] [CrossRef]
  86. Zhang, J.P.; Lencz, T.; Malhotra, A.K. D2 receptor genetic variation and clinical response to antipsychotic drug treatment: A meta-analysis. Am. J. Psychiatry 2010, 167, 763–772. [Google Scholar] [CrossRef] [PubMed]
  87. Ma, L.; Zhang, X.; Xiang, Q.; Zhou, S.; Zhao, N.; Xie, Q.; Zhao, X.; Zhou, Y.; Cui, Y. Association between dopamine receptor gene polymorphisms and effects of risperidone treatment: A systematic review and meta-analysis. Basic Clin. Pharmacol. Toxicol. 2019, 124, 94–104. [Google Scholar] [CrossRef] [PubMed]
  88. Lencz, T.; Robinson, D.G.; Napolitano, B.; Sevy, S.; Kane, J.M.; Goldman, D.; Malhotra, A.K. DRD2 promoter region variation predicts antipsychotic-induced weight gain in first episode schizophrenia. Pharmacogenet. Genom. 2010, 20, 569–572. [Google Scholar] [CrossRef]
  89. Bosia, M.; Lorenzi, C.; Pirovano, A.; Guglielmino, C.; Cocchi, F.; Spangaro, M.; Bramanti, P.; Smeraldi, E.; Cavallaro, R. COMT Val158Met and 5-HT1A-R -1019 C/G polymorphisms: Effects on the negative symptom response to clozapine. Pharmacogenomics 2015, 16, 35–44. [Google Scholar] [CrossRef]
  90. Hong, C.J.; Chen, T.J.; Yu, Y.W.; Tsai, S.J. Response to fluoxetine and serotonin 1A receptor (C-1019G) polymorphism in Taiwan Chinese major depressive disorder. Pharmacogenom. J. 2006, 6, 27–33. [Google Scholar] [CrossRef] [PubMed]
  91. Kato, M.; Fukuda, T.; Wakeno, M.; Okugawa, G.; Takekita, Y.; Watanabe, S.; Yamashita, M.; Hosoi, Y.; Azuma, J.; Kinoshita, T.; et al. Effect of 5-HT1A gene polymorphisms on antidepressant response in major depressive disorder. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2009, 150B, 115–123. [Google Scholar] [CrossRef]
  92. Lopez-Rodriguez, R.; Cabaleiro, T.; Ochoa, D.; Roman, M.; Borobia, A.M.; Carcas, A.J.; Ayuso, C.; Novalbos, J.; Abad-Santos, F. Pharmacodynamic genetic variants related to antipsychotic adverse reactions in healthy volunteers. Pharmacogenomics 2013, 14, 1203–1214. [Google Scholar] [CrossRef]
  93. Sanguesa, E.; Fernandez-Egea, E.; Concha, J.; Garcia, C.B.; Ribate, M.P. Impact of Pharmacogenetic Testing on Clozapine Treatment Efficacy in Patients with Treatment-Resistant Schizophrenia. Biomedicines 2024, 12, 597. [Google Scholar] [CrossRef]
  94. Blasi, G.; Selvaggi, P.; Fazio, L.; Antonucci, L.A.; Taurisano, P.; Masellis, R.; Romano, R.; Mancini, M.; Zhang, F.; Caforio, G.; et al. Variation in Dopamine D2 and Serotonin 5-HT2A Receptor Genes is Associated with Working Memory Processing and Response to Treatment with Antipsychotics. Neuropsychopharmacology 2015, 40, 1600–1608. [Google Scholar] [CrossRef]
  95. Basu, A.; Chadda, R.K.; Sood, M.; Kaur, H.; Kukreti, R. Association of serotonin transporter (SLC6A4) and receptor (5HTR1A, 5HTR2A) polymorphisms with response to treatment with escitalopram in patients with major depressive disorder: A preliminary study. Indian J. Med. Res. 2015, 142, 40–45. [Google Scholar] [CrossRef]
  96. Horstmann, S.; Lucae, S.; Menke, A.; Hennings, J.M.; Ising, M.; Roeske, D.; Muller-Myhsok, B.; Holsboer, F.; Binder, E.B. Polymorphisms in GRIK4, HTR2A, and FKBP5 show interactive effects in predicting remission to antidepressant treatment. Neuropsychopharmacology 2010, 35, 727–740. [Google Scholar] [CrossRef]
  97. Wu, Y.; Zeng, L.; Zhao, S. Ligands of Adrenergic Receptors: A Structural Point of View. Biomolecules 2021, 11, 936. [Google Scholar] [CrossRef]
  98. Zhang, F.; Steinberg, S.F. S49G and R389G polymorphisms of the beta(1)-adrenergic receptor influence signaling via the cAMP-PKA and ERK pathways. Physiol. Genom. 2013, 45, 1186–1192. [Google Scholar] [CrossRef]
  99. Parvez, B.; Chopra, N.; Rowan, S.; Vaglio, J.C.; Muhammad, R.; Roden, D.M.; Darbar, D. A common beta1-adrenergic receptor polymorphism predicts favorable response to rate-control therapy in atrial fibrillation. J. Am. Coll. Cardiol. 2012, 59, 49–56. [Google Scholar] [CrossRef]
  100. Pacanowski, M.A.; Gong, Y.; Cooper-Dehoff, R.M.; Schork, N.J.; Shriver, M.D.; Langaee, T.Y.; Pepine, C.J.; Johnson, J.A.; Investigators, I. beta-adrenergic receptor gene polymorphisms and beta-blocker treatment outcomes in hypertension. Clin. Pharmacol. Ther. 2008, 84, 715–721. [Google Scholar] [CrossRef]
  101. Sobczak, M.; Salaga, M.; Storr, M.A.; Fichna, J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: Current concepts and future perspectives. J. Gastroenterol. 2014, 49, 24–45. [Google Scholar] [CrossRef]
  102. Halikere, A.; Popova, D.; Scarnati, M.S.; Hamod, A.; Swerdel, M.R.; Moore, J.C.; Tischfield, J.A.; Hart, R.P.; Pang, Z.P. Addiction associated N40D mu-opioid receptor variant modulates synaptic function in human neurons. Mol. Psychiatry 2020, 25, 1406–1419. [Google Scholar] [CrossRef]
  103. Taqi, M.M.; Faisal, M.; Zaman, H. OPRM1 A118G Polymorphisms and Its Role in Opioid Addiction: Implication on Severity and Treatment Approaches. Pharmacogenom. Pers. Med. 2019, 12, 361–368. [Google Scholar] [CrossRef] [PubMed]
  104. Kusumi, I.; Boku, S.; Takahashi, Y. Psychopharmacology of atypical antipsychotic drugs: From the receptor binding profile to neuroprotection and neurogenesis. Psychiatry Clin. Neurosci. 2015, 69, 243–258. [Google Scholar] [CrossRef]
  105. Pan, Y.; Yao, J.; Wang, B. Association of dopamine D1 receptor gene polymorphism with schizophrenia: A meta-analysis. Neuropsychiatr. Dis. Treat. 2014, 10, 1133–1139. [Google Scholar] [CrossRef] [PubMed]
  106. Potkin, S.G.; Basile, V.S.; Jin, Y.; Masellis, M.; Badri, F.; Keator, D.; Wu, J.C.; Alva, G.; Carreon, D.T.; Bunney, W.E., Jr.; et al. D1 receptor alleles predict PET metabolic correlates of clinical response to clozapine. Mol. Psychiatry 2003, 8, 109–113. [Google Scholar] [CrossRef] [PubMed]
  107. Ota, V.K.; Spindola, L.N.; Gadelha, A.; dos Santos Filho, A.F.; Santoro, M.L.; Christofolini, D.M.; Bellucco, F.T.; Ribeiro-dos-Santos, A.K.; Santos, S.; Mari Jde, J.; et al. DRD1 rs4532 polymorphism: A potential pharmacogenomic marker for treatment response to antipsychotic drugs. Schizophr. Res. 2012, 142, 206–208. [Google Scholar] [CrossRef]
  108. de Matos, L.P.; Santana, C.V.; Souza, R.P. Meta-analysis of dopamine receptor D1 rs4532 polymorphism and susceptibility to antipsychotic treatment response. Psychiatry Res. 2015, 229, 586–588. [Google Scholar] [CrossRef] [PubMed]
  109. Shen, Y.C.; Chen, S.F.; Chen, C.H.; Lin, C.C.; Chen, S.J.; Chen, Y.J.; Luu, S.U. Effects of DRD2/ANKK1 gene variations and clinical factors on aripiprazole efficacy in schizophrenic patients. J. Psychiatr. Res. 2009, 43, 600–606. [Google Scholar] [CrossRef] [PubMed]
  110. Zhang, J.P.; Robinson, D.G.; Gallego, J.A.; John, M.; Yu, J.; Addington, J.; Tohen, M.; Kane, J.M.; Malhotra, A.K.; Lencz, T. Association of a Schizophrenia Risk Variant at the DRD2 Locus With Antipsychotic Treatment Response in First-Episode Psychosis. Schizophr. Bull. 2015, 41, 1248–1255. [Google Scholar] [CrossRef] [PubMed]
  111. Magistrelli, L.; Ferrari, M.; Furgiuele, A.; Milner, A.V.; Contaldi, E.; Comi, C.; Cosentino, M.; Marino, F. Polymorphisms of Dopamine Receptor Genes and Parkinson’s Disease: Clinical Relevance and Future Perspectives. Int. J. Mol. Sci. 2021, 22, 3781. [Google Scholar] [CrossRef]
  112. Zhou, W.; Xu, Y.; Lv, Q.; Sheng, Y.H.; Chen, L.; Li, M.; Shen, L.; Huai, C.; Yi, Z.; Cui, D.; et al. Genetic Association of Olanzapine Treatment Response in Han Chinese Schizophrenia Patients. Front. Pharmacol. 2019, 10, 177. [Google Scholar] [CrossRef]
  113. Naveen, M.; Patil, A.N.; Pattanaik, S.; Kaur, A.; Banerjee, D.; Grover, S. ABCB1 and DRD3 polymorphism as a response predicting biomarker and tool for pharmacogenetically guided clozapine dosing in Asian Indian treatment resistant schizophrenia patients. Asian J. Psychiatr. 2020, 48, 101918. [Google Scholar] [CrossRef]
  114. Zhou, W.; Chang, W.; Yan, Y.; Shen, L.; Li, W.; Yi, Z.; Qin, S. Pharmacogenetics analysis of serotonin receptor gene variants and clinical response to risperidone in Han Chinese schizophrenic patients. Neurosci. Lett. 2018, 683, 202–206. [Google Scholar] [CrossRef]
  115. Gupta, M.; Jain, S.; Moily, N.S.; Kaur, H.; Jajodia, A.; Purushottam, M.; Kukreti, R. Genetic studies indicate a potential target 5-HTR(3B) for drug therapy in schizophrenia patients. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2012, 159b, 1006–1008. [Google Scholar] [CrossRef]
  116. Melkersson, K.I.; Gunes, A.; Dahl, M.L. Impact of serotonin receptor 2A gene haplotypes on C-peptide levels in clozapine- and olanzapine-treated patients. Hum. Psychopharmacol. 2010, 25, 347–352. [Google Scholar] [CrossRef]
  117. Edinoff, A.N.; Akuly, H.A.; Hanna, T.A.; Ochoa, C.O.; Patti, S.J.; Ghaffar, Y.A.; Kaye, A.D.; Viswanath, O.; Urits, I.; Boyer, A.G.; et al. Selective Serotonin Reuptake Inhibitors and Adverse Effects: A Narrative Review. Neurol. Int. 2021, 13, 387–401. [Google Scholar] [CrossRef]
  118. Commons, K.G.; Linnros, S.E. Delayed Antidepressant Efficacy and the Desensitization Hypothesis. ACS Chem. Neurosci. 2019, 10, 3048–3052. [Google Scholar] [CrossRef]
  119. Chauhan, M.; Parry, R.; Bobo, W.V. Vilazodone for Major Depression in Adults: Pharmacological Profile and an Updated Review for Clinical Practice. Neuropsychiatr. Dis. Treat. 2022, 18, 1175–1193. [Google Scholar] [CrossRef] [PubMed]
  120. Zhao, X.; Huang, Y.; Li, J.; Ma, H.; Jin, Q.; Wang, Y.; Wu, L.; Zhu, G. Association between the 5-HT1A receptor gene polymorphism (rs6295) and antidepressants: A meta-analysis. Int. Clin. Psychopharmacol. 2012, 27, 314–320. [Google Scholar] [CrossRef] [PubMed]
  121. Dong, Z.Q.; Li, X.R.; He, L.; He, G.; Yu, T.; Sun, X.L. 5-HTR1A and 5-HTR2A genetic polymorphisms and SSRI antidepressant response in depressive Chinese patients. Neuropsychiatr. Dis. Treat. 2016, 12, 1623–1629. [Google Scholar] [CrossRef]
  122. Scutt, G.; Overall, A.; Scott, R.; Patel, B.; Hachoumi, L.; Yeoman, M.; Wright, J. Does the 5-HT(1A) rs6295 polymorphism influence the safety and efficacy of citalopram therapy in the oldest old? Ther. Adv. Drug Saf. 2018, 9, 355–366. [Google Scholar] [CrossRef] [PubMed]
  123. Bousman, C.A.; Stevenson, J.M.; Ramsey, L.B.; Sangkuhl, K.; Hicks, J.K.; Strawn, J.R.; Singh, A.B.; Ruano, G.; Mueller, D.J.; Tsermpini, E.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6, CYP2C19, CYP2B6, SLC6A4, and HTR2A Genotypes and Serotonin Reuptake Inhibitor Antidepressants. Clin. Pharmacol. Ther. 2023, 114, 51–68. [Google Scholar] [CrossRef]
  124. Wan, Y.-S.; Zhai, X.-J.; Tan, H.-A.; Ai, Y.-S.; Zhao, L.-B. Associations between the 1438A/G, 102T/C, and rs7997012G/A polymorphisms of HTR2A and the safety and efficacy of antidepressants in depression: A meta-analysis. Pharmacogenom. J. 2021, 21, 200–215. [Google Scholar] [CrossRef]
  125. Lin, J.Y.; Jiang, M.Y.; Kan, Z.M.; Chu, Y. Influence of 5-HTR2A genetic polymorphisms on the efficacy of antidepressants in the treatment of major depressive disorder: A meta-analysis. J. Affect. Disord. 2014, 168, 430–438. [Google Scholar] [CrossRef]
  126. Zhao, X.; Sun, L.; Sun, Y.H.; Ren, C.; Chen, J.; Wu, Z.Q.; Jiang, Y.H.; Lv, X.L. Association of HTR2A T102C and A-1438G polymorphisms with susceptibility to major depressive disorder: A meta-analysis. Neurol. Sci. 2014, 35, 1857–1866. [Google Scholar] [CrossRef] [PubMed]
  127. Yuan, R.; Yuan, F.; Ren, D.; Zhu, Y.; Bi, Y.; Hu, J.; Guo, Z.; Xu, F.; Niu, W.; Wu, X.; et al. HTR1A and HTR2A variants may not predict venlafaxine treatment response in China Han population with major depressive disorder. Psychiatry Res. 2018, 270, 1179–1180. [Google Scholar] [CrossRef] [PubMed]
  128. Mitsudomi, T.; Yatabe, Y. Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J. 2010, 277, 301–308. [Google Scholar] [CrossRef] [PubMed]
  129. Borgeaud, M.; Parikh, K.; Banna, G.L.; Kim, F.; Olivier, T.; Le, X.; Addeo, A. Unveiling the Landscape of Uncommon EGFR Mutations in NSCLC-A Systematic Review. J. Thorac. Oncol. 2024, 19, 973–983. [Google Scholar] [CrossRef] [PubMed]
  130. Bai, Y.; Liu, X.; Zheng, L.; Wang, S.; Zhang, J.; Xiong, S.; Zhang, P.; Jiao, Z.; Zhao, G.; Zhou, C.; et al. Comprehensive profiling of EGFR mutation subtypes reveals genomic-clinical associations in non-small-cell lung cancer patients on first-generation EGFR inhibitors. Neoplasia 2023, 38, 100888. [Google Scholar] [CrossRef]
  131. Zehir, A.; Benayed, R.; Shah, R.H.; Syed, A.; Middha, S.; Kim, H.R.; Srinivasan, P.; Gao, J.; Chakravarty, D.; Devlin, S.M.; et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 2017, 23, 703–713. [Google Scholar] [CrossRef]
  132. Wee, P.; Wang, Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers 2017, 9, 52. [Google Scholar] [CrossRef]
  133. Arkhipov, A.; Shan, Y.; Das, R.; Endres, N.F.; Eastwood, M.P.; Wemmer, D.E.; Kuriyan, J.; Shaw, D.E. Architecture and membrane interactions of the EGF receptor. Cell 2013, 152, 557–569. [Google Scholar] [CrossRef]
  134. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef]
  135. Amelia, T.; Kartasasmita, R.E.; Ohwada, T.; Tjahjono, D.H. Structural Insight and Development of EGFR Tyrosine Kinase Inhibitors. Molecules 2022, 27, 819. [Google Scholar] [CrossRef]
  136. Thomas, R.; Weihua, Z. Rethink of EGFR in Cancer With Its Kinase Independent Function on Board. Front. Oncol. 2019, 9, 800. [Google Scholar] [CrossRef] [PubMed]
  137. Uribe, M.L.; Marrocco, I.; Yarden, Y. EGFR in Cancer: Signaling Mechanisms, Drugs, and Acquired Resistance. Cancers 2021, 13, 2748. [Google Scholar] [CrossRef] [PubMed]
  138. Generali, D.; Leek, R.; Fox, S.B.; Moore, J.W.; Taylor, C.; Chambers, P.; Harris, A.L. EGFR mutations in exons 18–21 in sporadic breast cancer. Ann. Oncol. 2007, 18, 203–205. [Google Scholar] [CrossRef]
  139. Metzger, B.; Chambeau, L.; Begon, D.Y.; Faber, C.; Kayser, J.; Berchem, G.; Pauly, M.; Boniver, J.; Delvenne, P.; Dicato, M.; et al. The human epidermal growth factor receptor (EGFR) gene in European patients with advanced colorectal cancer harbors infrequent mutations in its tyrosine kinase domain. BMC Med. Genet. 2011, 12, 144. [Google Scholar] [CrossRef] [PubMed]
  140. Castaneda-Gonzalez, J.P.; Chaves, J.J.; Parra-Medina, R. Multiple mutations in the EGFR gene in lung cancer: A systematic review. Transl. Lung Cancer Res. 2022, 11, 2148–2163. [Google Scholar] [CrossRef]
  141. Abourehab, M.A.S.; Alqahtani, A.M.; Youssif, B.G.M.; Gouda, A.M. Globally Approved EGFR Inhibitors: Insights into Their Syntheses, Target Kinases, Biological Activities, Receptor Interactions, and Metabolism. Molecules 2021, 26, 6677. [Google Scholar] [CrossRef]
  142. Nagasaka, M.; Zhu, V.W.; Lim, S.M.; Greco, M.; Wu, F.; Ou, S.I. Beyond Osimertinib: The Development of Third-Generation EGFR Tyrosine Kinase Inhibitors For Advanced EGFR+ NSCLC. J. Thorac. Oncol. 2021, 16, 740–763. [Google Scholar] [CrossRef]
  143. Hanley, M.J.; Camidge, D.R.; Fram, R.J.; Gupta, N. Mobocertinib: Mechanism of action, clinical, and translational science. Clin. Transl. Sci. 2024, 17, e13766. [Google Scholar] [CrossRef]
  144. Cai, W.Q.; Zeng, L.S.; Wang, L.F.; Wang, Y.Y.; Cheng, J.T.; Zhang, Y.; Han, Z.W.; Zhou, Y.; Huang, S.L.; Wang, X.W.; et al. The Latest Battles Between EGFR Monoclonal Antibodies and Resistant Tumor Cells. Front. Oncol. 2020, 10, 1249. [Google Scholar] [CrossRef]
  145. PharmGKB Database. EGFR. Clinical Annotations. Available online: https://www.pharmgkb.org (accessed on 20 June 2024).
  146. Costa, D.B. Kinase inhibitor-responsive genotypes in EGFR mutated lung adenocarcinomas: Moving past common point mutations or indels into uncommon kinase domain duplications and rearrangements. Transl. Lung Cancer Res. 2016, 5, 331–337. [Google Scholar] [CrossRef]
  147. Yasuda, H.; Park, E.; Yun, C.H.; Sng, N.J.; Lucena-Araujo, A.R.; Yeo, W.L.; Huberman, M.S.; Cohen, D.W.; Nakayama, S.; Ishioka, K.; et al. Structural, biochemical, and clinical characterization of epidermal growth factor receptor (EGFR) exon 20 insertion mutations in lung cancer. Sci. Transl. Med. 2013, 5, 216ra177. [Google Scholar] [CrossRef]
  148. Castellanos, E.; Feld, E.; Horn, L. Driven by Mutations: The Predictive Value of Mutation Subtype in EGFR-Mutated Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 612–623. [Google Scholar] [CrossRef] [PubMed]
  149. Oliveira-Cunha, M.; Newman, W.G.; Siriwardena, A.K. Epidermal growth factor receptor in pancreatic cancer. Cancers 2011, 3, 1513–1526. [Google Scholar] [CrossRef] [PubMed]
  150. Ma, X.; Liu, X.; Ou, K.; Zhang, M.; Gao, L.; Yang, L. Advanced pancreatic cancer with KRAS wild-type and EGFR-sensitive mutation respond favorably to furmonertinib: A case report. Front. Oncol. 2023, 13, 1151178. [Google Scholar] [CrossRef]
  151. Mody, J.; Kamgar, M. Pancreatic Adenocarcinoma with Co-Occurrence of KRAS and EGFR Mutations: Case Report and Literature Review. Case Rep. Oncol. 2024, 17, 399–406. [Google Scholar] [CrossRef] [PubMed]
  152. Doleschal, B.; Petzer, A.; Rumpold, H. Current concepts of anti-EGFR targeting in metastatic colorectal cancer. Front. Oncol. 2022, 12, 1048166. [Google Scholar] [CrossRef]
  153. Garcia-Foncillas, J.; Sunakawa, Y.; Aderka, D.; Wainberg, Z.; Ronga, P.; Witzler, P.; Stintzing, S. Distinguishing Features of Cetuximab and Panitumumab in Colorectal Cancer and Other Solid Tumors. Front. Oncol. 2019, 9, 849. [Google Scholar] [CrossRef] [PubMed]
  154. Taberna, M.; Oliva, M.; Mesia, R. Cetuximab-Containing Combinations in Locally Advanced and Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma. Front. Oncol. 2019, 9, 383. [Google Scholar] [CrossRef]
  155. Rehmani, H.S.; Issaeva, N. EGFR in head and neck squamous cell carcinoma: Exploring possibilities of novel drug combinations. Ann. Transl. Med. 2020, 8, 813. [Google Scholar] [CrossRef]
  156. Gadgeel, S.M.; Chen, W.; Cote, M.L.; Bollig-Fischer, A.; Land, S.; Schwartz, A.G.; Bepler, G. Fibroblast growth factor receptor 1 amplification in non-small cell lung cancer by quantitative real-time PCR. PLoS ONE 2013, 8, e79820. [Google Scholar] [CrossRef] [PubMed]
  157. Vouri, M.; Croucher, D.R.; Kennedy, S.P.; An, Q.; Pilkington, G.J.; Hafizi, S. Axl-EGFR receptor tyrosine kinase hetero-interaction provides EGFR with access to pro-invasive signalling in cancer cells. Oncogenesis 2016, 5, e266. [Google Scholar] [CrossRef] [PubMed]
  158. Ma, Y.; Tang, N.; Thompson, R.C.; Mobley, B.C.; Clark, S.W.; Sarkaria, J.N.; Wang, J. InsR/IGF1R Pathway Mediates Resistance to EGFR Inhibitors in Glioblastoma. Clin. Cancer Res. 2016, 22, 1767–1776. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pharmacogenetics overview. Traditional medicine operates based on the “one drug fits all” paradigm, treating all patients identically, while precision medicine employs “omics” methods, mainly pharmacogenomics, with the aim of providing patients with personalized treatment. Genetic differences between individuals may arise from different types of mutations, with the most prevalent being SNPs. In order to establish a correlation between a genetic variant and drug response, two methods are currently performed: candidate gene association studies and genome-wide association studies, each with its own advantages [21,22,23]. Adapted from “Personalized Medicine (AI vs. Traditional Techniques)” and “The Principle of a Genome-wide Association Study (GWAS)” template by BioRender.com. Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 March 2024).
Figure 1. Pharmacogenetics overview. Traditional medicine operates based on the “one drug fits all” paradigm, treating all patients identically, while precision medicine employs “omics” methods, mainly pharmacogenomics, with the aim of providing patients with personalized treatment. Genetic differences between individuals may arise from different types of mutations, with the most prevalent being SNPs. In order to establish a correlation between a genetic variant and drug response, two methods are currently performed: candidate gene association studies and genome-wide association studies, each with its own advantages [21,22,23]. Adapted from “Personalized Medicine (AI vs. Traditional Techniques)” and “The Principle of a Genome-wide Association Study (GWAS)” template by BioRender.com. Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 March 2024).
Ijms 25 09371 g001
Figure 2. GPCR signaling and functional consequences of mutated forms. Upon ligand binding, GPCRs function like GEFs (guanine -exchange factors) favoring the exchange of GDP-bound Gα subunit to GTP-bound Gα subunits, initiating the signaling cascade. Mutations present in the ligand binding pocket (different for every GPCR class [72]) will halt the ligand–receptor interaction. Genetic variants affecting the G protein/β-arrestin binding domains (ICL2, ICL3, and C-tail [61,73]) can result in blocked or biased signaling. Moreover, some mutated receptor forms can present altered trafficking and disturbed desensitization, internalization, and degradation mechanisms [74]. Adapted from “Cell Endocytosis (Layout)” by BioRender.com. Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 March 2024).
Figure 2. GPCR signaling and functional consequences of mutated forms. Upon ligand binding, GPCRs function like GEFs (guanine -exchange factors) favoring the exchange of GDP-bound Gα subunit to GTP-bound Gα subunits, initiating the signaling cascade. Mutations present in the ligand binding pocket (different for every GPCR class [72]) will halt the ligand–receptor interaction. Genetic variants affecting the G protein/β-arrestin binding domains (ICL2, ICL3, and C-tail [61,73]) can result in blocked or biased signaling. Moreover, some mutated receptor forms can present altered trafficking and disturbed desensitization, internalization, and degradation mechanisms [74]. Adapted from “Cell Endocytosis (Layout)” by BioRender.com. Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 March 2024).
Ijms 25 09371 g002
Figure 3. EGFR mutations and signaling in cancer. Kinase-activating mutations and EGFR overexpression are responsible for EGFR signaling amplification, which defines the typical cancer cell: uncontrolled proliferation and survival. EGFR-targeted therapy improves patient prognosis. Nonetheless, secondary mutations can emerge (T790M and C797S, in bold), giving rise to resistant cancer cells. Adapted from “EGFR and EGFRvIII Roles in Cancer Biology” by BioRender.com. Retrieved from https://app.biorender.com/biorender-templates, (accessed on 28 March 2024) [134,135].
Figure 3. EGFR mutations and signaling in cancer. Kinase-activating mutations and EGFR overexpression are responsible for EGFR signaling amplification, which defines the typical cancer cell: uncontrolled proliferation and survival. EGFR-targeted therapy improves patient prognosis. Nonetheless, secondary mutations can emerge (T790M and C797S, in bold), giving rise to resistant cancer cells. Adapted from “EGFR and EGFRvIII Roles in Cancer Biology” by BioRender.com. Retrieved from https://app.biorender.com/biorender-templates, (accessed on 28 March 2024) [134,135].
Ijms 25 09371 g003
Table 1. Known GPCR variants that affect drug efficacy and/or safety. LABA, long-acting β-agonists; * first generation: chlorpromazine and haloperidol; second generation: clozapine, risperidone, olanzapine, and aripiprazole.
Table 1. Known GPCR variants that affect drug efficacy and/or safety. LABA, long-acting β-agonists; * first generation: chlorpromazine and haloperidol; second generation: clozapine, risperidone, olanzapine, and aripiprazole.
GeneVariantLigandsEffectsRef.
ADRB1rs1801253:R389GMetoprolol
Carvedilol
Bisoprolol
↓ Antihypertensive response[76,77,78]
rs1801252:S49GAtenolol↑ Adverse cardiovascular events[79]
ADRB2rs1042713:R16GLABA↑ Risk of asthma exacerbation[80]
rs1800888:T164ISalbutamol↓ Ligand binding[81]
ADRA2Crs11269124: Del 322–325Clonidine↑ Efficacy[82]
OPM1rs1799971:N40DEndogenous opioids
Buprenorphine
↑ β-endorphin binding affinity
↓ Expression
↓ Efficacy for buprenorphine
[83,84,85]
DRD2rs1799732
DRD2 PROM −141C Ins/Del
Antipsychotics *↓ Efficacy
↑ Weight gain
[86,87,88]
HTR1Ars6295Clozapine↑ Efficacy[89]
Fluoxetine
Paroxetine
Milnacipran
↑ Efficacy[90,91]
HTR2Ars6313Risperidone↑ Adverse cardiovascular events[92]
rs6314Clozapine↑ Efficacy[93]
Olanzapine↑ Efficacy[94]
rs6311Escitalopram↑Memory loss[95]
rs7997012Antidepressants↑ Treatment outcome
↑ Remission
[96]
Table 2. TKIs and monoclonal antibodies that target EGFR based on the FDA Table of Pharmacogenomic Biomarkers in Drug Labeling [13]. NSCLC, non-small-cell lung cancer; CRC, colorectal cancer; and SCCHN, squamous cell carcinoma of the head and neck.
Table 2. TKIs and monoclonal antibodies that target EGFR based on the FDA Table of Pharmacogenomic Biomarkers in Drug Labeling [13]. NSCLC, non-small-cell lung cancer; CRC, colorectal cancer; and SCCHN, squamous cell carcinoma of the head and neck.
DrugDrug ClassLabeling SectionsType of CancerTargeted Mutation
ErlotinibTKI (I)Indications and usage, dosage and administration, adverse reactions, and clinical studiesNSCLC
Pancreatic cancer
Exon 19 deletion
Exon 21 L858R
GefitinibTKI (I)Indications and usage, dosage and administration, and clinical studiesNSCLCExon 19 deletion
Exon 21 L858R
AfatinibTKI (II)Indications and usage, dosage and administration, adverse reactions, and clinical studiesNSCLCExon 19 deletion
Exon 21 L858R
DacomitinibTKI (II)Indications and usage, dosage, and administration, adverse reactions, use in specific populations, and clinical studiesNSCLCExon 19 deletion
Exon 21 L858R
OsimertinibTKI (III)Indications and usage, dosage and administration, adverse reactions, and clinical studiesNSCLCExon 19 deletion
Exon 21 L858R
T790M
MobocertinibTKIIndications and usage, dosage and administration, adverse reactions, and clinical studiesNSCLCExon 20 insertion
Amivantamab-vmjwMabIndications and usage, dosage and administration, adverse reactions, and clinical studiesNSCLCExon 20 insertion
Cetuximab MabIndications and usage, dosage and administration, adverse reactions, and clinical studiesCRC
SCCHN
PanitumumabMabAdverse reactions, clinical pharmacology, and clinical studiesCRC
Table 3. Clinical EGFR-related annotations from the PharmGKB database [145].
Table 3. Clinical EGFR-related annotations from the PharmGKB database [145].
DrugEGFR VariantLevel of EvidencePharmacologyPhenotype
Gefitinibrs1214345681AEfficacyCarcinoma, NSCLC
rs1214345692BEfficacyCarcinoma, NSCLC, drug resistance
rs22933473EfficacyCarcinoma, NSCLC
rs7128293EfficacyNeoplasms
rs115683153EfficacyCarcinoma, NSCLC
Erlotinibrs1214345692BEfficacyAdenocarcinoma, carcinoma, NSCLC, drug resistance, lung neoplasms
rs7128293EfficacyNeoplasms
rs7128293ToxicityNeoplasms
rs22279833ToxicityCarcinoma, NSCLC, colorectal neoplasms, neoplasms, pancreatic neoplasms
Cetuximabrs22279833EfficacyHead and neck neoplasms
rs7128293EfficacyColorectal neoplasms
rs7128303Toxicity Colorectal neoplasms
Panitumumabrs7128293EfficacyColorectal neoplasms
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Anghel, S.A.; Dinu-Pirvu, C.-E.; Costache, M.-A.; Voiculescu, A.M.; Ghica, M.V.; Anuța, V.; Popa, L. Receptor Pharmacogenomics: Deciphering Genetic Influence on Drug Response. Int. J. Mol. Sci. 2024, 25, 9371. https://doi.org/10.3390/ijms25179371

AMA Style

Anghel SA, Dinu-Pirvu C-E, Costache M-A, Voiculescu AM, Ghica MV, Anuța V, Popa L. Receptor Pharmacogenomics: Deciphering Genetic Influence on Drug Response. International Journal of Molecular Sciences. 2024; 25(17):9371. https://doi.org/10.3390/ijms25179371

Chicago/Turabian Style

Anghel, Sorina Andreea, Cristina-Elena Dinu-Pirvu, Mihaela-Andreea Costache, Ana Maria Voiculescu, Mihaela Violeta Ghica, Valentina Anuța, and Lăcrămioara Popa. 2024. "Receptor Pharmacogenomics: Deciphering Genetic Influence on Drug Response" International Journal of Molecular Sciences 25, no. 17: 9371. https://doi.org/10.3390/ijms25179371

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop