Companion diagnostics (CDx) is a biomarker assay linked to a specific drug. It is a drug-diagnostic codeveloped model defining the probable benefit of a drug with a molecular diagnostic assay (Jørgensen 2016). CDx assay is an imperative decision tool for a pharmacotherapeutic intervention, and an incorrect result may lead to an inappropriate treatment. The assays have an essential role in drug development process, and the efficiency depends on the performance. On approval of a drug, molecular diagnostic assays are regarded as a companion to drug, and hence named companion diagnostics. CDx is important both in drug development and individualized treatment (Figure 2).
In the drug-diagnostic co-development for CDx assays, the critical factors are biomarker hypothesis, analytical validation, and clinical relevance. A biomarker hypothesis is developed from a complete data on the pathophysiology and drug mechanism (Jørgensen and Nielsen 2017). The selection of assay method depends on biomarker type. The assay sensitivity and specificity should be relevant to the sample (Jørgensen 2020). With the development of prototype for assay, analytical verification is done to identify the stability, feasibility, and reproducibility. Then, the prototype is employed in an preliminary clinical trial to correlate the biomarker with clinical outcome. The outcome data is used to select the cutoff that defines the positive test result, later transforming as a decision tool. For example, Trastuzumab (HERCEPTIN) improves survival in both adjuvant and metastatic HER2-positive breast cancer in patients (Hudis 2007). Thus, the HER2 test became the first CDx, and additional assays were then approved by FDA as CDx later. However, the HER2 assays were not codeveloped with trastuzumab, but were instead approved after the initial drug approval as technologies and commercial opportunities evolved (Scheerens, Malong et al. 2017). Another example is the COBAS BRAF V600E test. FDA approved this test along with vemurafenib (ZELBORAF) for metastatic melanoma in which the overall survival was improved in patients with the BRAF V600E mutation compared to the control drug (Chapman, Hauschild et al. 2011).
Plotting true positivity (sensitivity) against false positive (specificity) is the method used to determine the preliminary cutoffs. The area under the curve (AUC) is calculated for the different cutoff points and the one giving the largest AUC is selected as the final clinical cutoff for the assay with values ranging between 0.50 and 1.0(Nahm 2022). A value of 0.5 indicates no discrimination and 1.0 indicates perfect discrimination and a value of ≥ 0.8 is suggests an excellent biomarker. The AUC of the ROC curve reflects the overall accuracy and separation performance of the biomarker (or biomarkers) and can be readily used to compare different biomarker combinations or models (Šimundić 2009) (Sanghera, Orlando et al. 2013).
The analytical validation is complete with the selection of cutoff for the CDx. A CDx assay developed within a single laboratory is a laboratory-developed test (LDT). The final step is to determine the external reproducibility study among the clinical laboratories (Meinert, Alturkistani et al. 2019). Clinical validation is not initiated before analytical validation including sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV).
Clinical validation may include both randomized and nonrandomized trial in which the intrapopulation variability is decreased and proportion of responsive patients increased by preselection (Chapman, Hauschild et al. 2011). The drug-diagnostic co-development is superior to the traditional all-comers approach (Jørgensen 2015). Drugs in the pipelines mostly are developed from molecular subsets of patients relying on biomarker selection (Milne, Cohen et al. 2015). Traditional methods of drug development are replaced by the adaptive development approaches that are efficacious in a relatively small specific patient population. For instance, in case of oncology drug development whether or not they have a CDx assay linked to their use, there is a clear higher objective response rates for the group of drugs with a CDx from 80.2% to 41.0%, while for the group of drugs with no CDx assay linked to use from 45.0% to 6.8% (Jørgensen and Nielsen 2017).
Antimicrobials are designed in an “one-size-fits-all” approach (Geli, Laxminarayan et al. 2012). Clinical microbiology counts on the in vitro analysis to guide diagnosis and therapeutic efficacy (Jørgensen and Nielsen 2017, Jørgensen 2020). The pharmacodynamic mechanisms are distinct for each of the antimicrobial agents. Since the 1950s, there has been a decline in drug introduction into the market with a success rate of only 3% derived from conventional methods. However, a genetic based authorization accounts for 50% of all marketed drugs to be efficacious. The advantage of CDx is that it improves prognosis, predictive response and tolerance to treatment (Meinert, Alturkistani et al. 2019). However, no specific CDx prescribed for infectious diseases have been approved by rhe FDA.
As mentioned, principal objectives of CDx is identification of appropriate patient groups, therapeutic product, prediction of adverse reactions, monitor response, adjust the dosage scheme and ensure improved treatment outcome (Kaufmann, Keppens et al. 2015). This coordination of drug and CDx in infectious diseases needs to be further addressed. Primarily the important facets to be considered in devising the strategy for CDx are – the analyte and assay, the drug and target. The nature of the antimicrobial agent to be employed and the pathogen against which it is targeted are fundamental in CDx paradigm. With an expansion in the spectrum of antimicrobial agents, there is an unprecedented array for potential drug selection. Protocols need to be implemented to ensure specificity of therapies and their effects on direct and collateral targets. The drug-and-target-focused approach is envisioned to be braced by artificial intelligence and machine learning technologies.
Common barrier in implementation of CDx clinically is the delay in adoption of assay due to lack of awareness on clinical relevance. Other factors in play are accessibility, availability, quality of sampling and inaccuracy from insufficient amount and low quality. Lack of testing in the existing labs, false negatives and false positives reporting may also mislead treatment decisions. Further, the accuracy of test, turnaround time and its interaction also impact CDx development.
In case of complementary diagnostics, companion diagnostics restrict patients to receive co-developed therapies based on the outcome emphasizing the biomarker.
CDx with alternate therapies for infectious diseases
Antibacterial mAbs are making a comeback as the phage therapy (Kaplon and Reichert 2019). With the FDA approving 4 mAbs for pathogens, the key factor influencing its development in CDx is finding the optimal targets for the pathogen and isotype(Wang-Lin and Balthasar 2018). Antitoxin antibodies as preventive strategy or as adjunctive to antibiotics have yielded success (Aguilar, Varshney et al. 2017, (Motley, Banerjee et al. 2019). Other antibodies targeting the surface proteins - outer membrane proteins involved in adhesion , immune evasion and bacterial biosynthesis may be strategized for CDx biomarker development (Visan, Rouleau et al. 2018, Ali, Yu et al. 2019) (Chen, Sun et al. 2019) (Varshney, Kuzmicheva et al. 2018).
Polysaccharides, namely lipopolysaccharide (LPS) and capsular polysaccharide (CPS) are popular targets. CPS targeted antibodies improve opsonophagocytosis (Kobayashi, Porter et al. 2018). A successful mAb therapy against a polysaccharide antigen shifts bacterial populations away from utilizing that antigen (Doyle, Moon et al. 2018) and hence may be utilized in the identification with IHC or fluorescence activated cell sorter FACS. Natural LPS antibodies, have high frequency of somatic hypermutations in IgM and IgA against certain glycan signatures that significantly improve the affinity, specificity, and stability of several preclinical antibodies with an application of adding multi-specific binding properties(Rollenske, Szijarto et al. 2018).
Three anti-bacterial mAb products based on neutralization of exotoxins have been approved for human use - Raxibacumab (ABthrax®), Obiltoxaximab (Anthim®) as a prophylactic for anthrax, and Bezlotoxumab (ZINPLAVATM) for recurrent infection by Clostridum difficile. Antibiotics might control the bacterial infection but fail to clear released toxins from the bloodstream. The detection of toxin in the blood may be neutralized with Raxibacumab, the anti-PA recombinant, a IgG1λ mAb, and prevent disease progression. Serodiagnostic assays for anthrax are based on detection of antibodies against PA or lethal factor (LF) (Ghosh and Goel 2012, Ghosh, Tomar et al. 2013), which develop post infection. However, early detection by sandwich ELISA 24 or surface plasmon resonance 8 could provide a timely diagnosis. Hence, either of the detection methods along with Raxibacumab (ABthrax®), Obiltoxaximab (Anthim®) may be prescribed as a CDx for anthrax.
Known as host defense peptides (HDP), AMPs exhibit antimicrobial activity on both Gram-negative and gram-positive bacteria and belong to two main families - the defensins and cathelicidins. Antimicrobial peptides are small peptides (4–50 amino acid residues) with amphipathic conformation (Mercer, Stewart et al. 2019, Torres, Sothiselvam et al. 2019). The protective effect of AMP against infections has clinical correlations (de la Fuente-Núñez, Silva et al. 2017, Coates, Blanchard et al. 2018). In patients, with impaired epithelial AMP production as in the case of atopic dermatitis susceptibility to secondary infections are more as in contrast to conditions with increased AMP production (e.g., psoriasis) (Ong, Ohtake et al. 2002). Hence, assessment of AMP induction by IHC, Western blot, RT-PCR may be employed in the diagnostic part of AMP CDx.
In alternate measure, AMPs have significant efficacy in preventing biofilm formation, despite the heterogenous nature and complexity of biofilm (Haney, Trimble et al. 2018, Magana, Pushpanathan et al. 2020). AMP activity on biofilm is best described by growing in multi-well plates or the Calgary device. Fulfilling the requirements for a CDx, criterion of an in vitro diagnostics IVD, the Calgary device, a flow-cell device, may be combined with a AMP assay(Macià, Rojo-Molinero et al. 2014). In the case of membrane-active AMPs, membrane permeabilization effects although fluorescence microscopy with fluorophores are used, an inexpensive methods is the microtiter plates for detection with Crystal violet (O'Toole 2011).
Phage therapies are unlikely be the first line treatment but can be an alternative in cases that have failed with antibiotic treatments (Pires, Costa et al. 2020). Phage preparations can be formulated if the preliminary pathogenic profile is known. Both phages and bacteria are subject to continuous co-evolution (Dion, Oechslin and Moineau 2020). Phage therapy has emerged as a potential alternative with success, and one that meets the One Health Approach with European Green Deal (European Commission 2019). However, complicating regulatory issues and safety concerns prevent in phage in therapeutics (Chanishvili, 2012). However, use of phages with antibiotics is a superior strategy for controlling bacterial pathogens with a dual approach of stronger bacterial suppression and the reduced capacity for developing phage and/or antibiotic resistance (Torres-Barceló and Hochberg 2016). Phage productivity and phage-mediated bacterial lysis with PAS is beneficial for some phage/antibiotic combinations, but ineffective in others (Uchiyama, Shigehisa et al. 2018, Gelman, Beyth et al. 2018, Torres-Barceló, Gurney et al. 2018). A combined approach restores antibiotic sensitivity (Chan et al., 2016). The diverse properties are not exploited in phage-antibiotic combinations. Depending on the magnitude of bacterial suppression, the interactions are categorized as true synergism, additive effects, or as facilitation (Chaudhry, Concepción-Acevedo et al. 2017).
Recently phage susceptibility test has been developed to simultaneously test hundreds of phages selected from Adaptive Phage Therapeutic’s (APT) phage bank, against bacteria isolated from a patient. The PST identifies one or more phage for treatment. Phage library, the APT’s phage bank, has been deployed with a companion diagnostic to achieve rapid response and cost-effective therapy for otherwise recalcitrant bacterial infections. (APT,2017). Apart from natural phages, synthetic phages with engineered genes can be employed. Minimal phage cassis can replicate well in a wide range of target bacteria is the target. Hybrid phages have interchangeable tails, with lower percentage of homology, and adaptable to target bacteria. For selectivity, this is combined with the receptor binding proteins (RBPs) selected by in vitro evolution in lab, and determines the strain to be killed by synthetic phage. Multivalent phages with multiple RBPs can expand the host range, if the therapeutic phage is intended to be used on a wider range of bacteria.
With regards to aptamers, recognition and specific binding are promising aspects. Selection of aptamers from oligonucleotide pools by SELEX can allow a wide range of biomedically relevant targets (Tan, Donovan et al. 2013). From well-established selection process, new aptamers can be selected for the target particularly biomarkers like kinases, growth factors, and cell-surface receptors (Xing, Hwang et al. 2014). Notably, the use of DNA aptamers for diagnosis and targeted therapy is forthcoming in CDx. The incorporation of DNA aptamers into CDx is hugely untapped. Therapeutically, nanomaterials like liposomes, polymer vesicles and silica nanoparticles, combined with DNA aptamers can be used for in vivo targeted drug delivery with physiochemical stability, stimuli-responsiveness, controlled release profile and desired in vivo biodistribution. It has long been recognized as favorable candidate in theranostics-combination of diagnosis and therapy in one system, its therapeutic side may hold the key to success in AMR. With identification of markers from infected patients, expedition of in vitro selection of DNA aptamers facilitates the production of therapeutic tool designed specifically.
Cao et al 2009 combined a panel of ssDNA aptamers in the detection of S.aureus with five ssDNA aptamers capable of binding to different protein targets that detects in pyogenic fluid of burn victims. Conjugation of these aptamers to the surface of single-walled carbon nanotubes (SWCNTs) detected S.aureus via real-time potentiometry with high sensitivity (Zelada-Guillén, Sebastián-Avila et al. 2012).
Aptamers and CRISPR-Cas are powerful diagnostic and therapeutic tool for AMR crisis (Kaur, Bruno et al. 2018). The extra cellular and intracellular targets for aptamers are huge and can be used to treat resistant bacteria. Aptamers are chemical antibodies (Toh, Citartan et al. 2015) binding at upto 1 pM affinity (Chen, Rashid et al. 2015, Ha, Jung et al. 2017) and sensitive to single amino acid mutation Owing to their small size, they have better reach accessibility that is otherwise inaccessible mAbs (Xiang, Zheng et al. 2015).
One of requisite for CDx is an in vitro diagnostic device. Different diagnostic devices that are aptamer based include Aptamer-Linked Immobilized Sorbent Assay (ALISA), dot-blot, lateral-flow strips conjugated to nanomaterials, and aptamer-based sensors (Stoltenburg, Krafčiková et al. 2016, Xiong, Zhang et al. 2020). DNA aptamers detected purified PBP2a protein (Fan, Cui et al. 2020) as well in clinical sample (Qiao, Meng et al. 2018).
With respect to the assay different formats of detection with aptamers are Enzyme-Linked Oligonucleotide Assay (ELONA), fluorescence-based assay, aptamer-based flow cytometry, fluorogenic assay and electrochemical sensing are promising diagnostic tool. The next criterion in CDx, which is the biomarker development can be accomplished by the highly multiplexed SOMAmer (Slow Off-rate Modified Aptamer)-based biomarker discovery. Diagnostic platform-SOMAscan detects and quantifies >1,300 proteins simultaneously in a variety of clinical sample (Candia, Cheung et al. 2017). In one instance, SOMAscan discovered several biomarkers (Russell, Green et al. 2017).
The other tool is CRISPR-CAS (clustered repetitive interspaced short palindromic repeats, CRISPR-associated enzyme) against antimicrobial-resistant pathogens. Diagnostic platforms use Cas enzymes (Cas12/Cas13) incubated with the target nucleic acid and fluorescent ssDNA/ssRNA reporters. On detection of target, the Cas enzymes trans-cleave and generate a robust fluorescent signal that has been correlated with PCR-based methods (Gootenberg, Abudayyeh et al. 2018). A different multiplexing strategy also uses Cas9 to enrich low-abundance targets from complex backgrounds before NGS. This method aid in distinguishing Klebsiella pneumoniae carbapenemase (KPC) and New Delhi metallo-β-lactamase (NDM) from five clinical isolates of K. pneumoniae (Gootenberg et al., 2017)
CRISPR-Cas9, Cas3, and Cas13 are the potent sequence-specific antimicrobial. CRISPR interference (CRISPRi) uses catalytically inactive Cas9 (dCas9) and single-guide RNA (sgRNA) to repress sequence-specific genes (Zhang, Xu et al. 2021). Cell death occurs when single guide RNA is directed to genes on the chromosome or plasmids containing a toxin–antitoxin system. In the absence of toxin–antitoxin, plasmid clearance or drastic reduction of copy number is achieved (Bikard, Euler et al. 2014, Kiga, Tan et al. 2020) (Kiga, Tan et al. 2020) (Tagliaferri, Guimarães et al. 2020).
Recently, recognition of surface proteins on methicillin resistant staphylococcus aureus (MRSA) strains by aptamer and CRISPR-Cas12a-assisted rolling circle amplification (Xu, Dai et al. 2020). There is an arcade of CRISPR-Cas/aptamer combinations and target bacteria to be tested. CRISPR-Cas and aptamers can be combined to treat and/or diagnose resistant bacterial infections due to their aforementioned characteristics, making a pair of a companion test. Exogenous short interfering RNA (siRNA) alters gene expression but exhibit high stability with minimal toxicity, modulating virulence, drug resistance and pathogenesis. The siRNA–aptamer conjugates increased therapeutic efficacy and safety (Afrasiabi, Pourhajibagher et al. 2020). As a result, a companion test may include siRNA-aptamer combination.