Autore/Fonte: Nat Commun 14, 6756 (2023)
Cancro ovarico: l’intelligenza artificiale prevede le risposte terapeutiche
Leggi →
14 Novembre 2023
Questo è quello che abbiamo trovato per te
Autore/Fonte: Nat Commun 14, 6756 (2023)
La conferma da uno studio svolto in Finlandia
Circulation, Volume 148, Issue Suppl_1, Page A16919-A16919, November 6, 2023. PAMAM dendrimers are 3-dimensional nanomolecules which have the capability to deliverbiomolecules and gene therapy to the cellsinvitroandinvivo. We have previously used the generation-4 surface-modified PAMAM dendrimers having 90% hydroxyl groups and 10% amine groups (G4-90/10) which are safe, cross the blood-brain barrier and deliver the biomolecules into the cells. In thecurrent study, we utilized G4-90/10 PAMAM dendrimers to deliver a gene therapy to the infarct in aischemic stroke brain in rats. Following an ischemic stroke, glial cells (such as astrocytes) are recruitedat the infarct region to perform maintenance functions, creating severe neuroinflammation. The goalof this study was to reprogram some of the astrocytes into neuronal lineage and repopulate the infarctregion. In addition, we also analyzed the temporal profile of astrocyte activation following ischemicstroke in the striatal and cortical regions to determine the time-point at which the maximum gliosisoccurs. This gives us a crucial timeline for treatment intervention to achieve astrocytereprogramming. We have used a transcription factor (hSOX2 gene), which was then delivered to thestroke brain in rats using the G4-90/10 dendrimers. Our results showed that the maximum glial cellactivation occurs in striatal and cortical regions on day 4 following ischemic stroke. Following genetherapy intervention, our initial analyses indicated that there was recovery from behavioral deficits inthe stroked rats compared to the vehicle-treated stroked rats and sham controls. Overall, these resultsshow that PAMAM dendrimers have significant potential delivering therapeutic molecules for treatingstroke and other neurological conditions.Support for this study was provided by the American Heart Association (18AIREA 33990094),Neuroscience program, the College of Medicine, department of Chemistry and Biochemistry, the JohnG. Kulhavi Professorship and the E. Malcolm Field and Gary Leo Dunbar Endowed Chair of Neuroscience at Central Michigan University.
Circulation, Volume 148, Issue Suppl_1, Page A12927-A12927, November 6, 2023. Background:Hypertrophic cardiomyopathy (HCM) is a life-threatening inherited heart disease characterized by left ventricular hypertrophy and diastolic dysfunction. The most common cause of HCM is genetic variants inMYBPC3, encoding cardiac myosin binding protein C (cMyBP-C), a sarcomeric protein with structural and regulatory roles. The majority ofMYBPC3gene variants are truncating leading to protein haploinsufficiency.Hypothesis:Transfer of a functional copy ofMYBPC3to heart muscle deficient in cMyBP-C will lead to sustained improvements in cardiac function.Aims:To determine whether BMN 293 (AAV-hMYBPC3) can restore cMyBP-C levels in the sarcomere and halt and/or reverse disease progression in non-clinical models of genetic HCM due toMYPBC3deficiency.Methods:BMN 293 is an adeno-associated virus (AAV) vector that encodes wild-type humanMYBPC3under the control of a cardiomyocyte-selective promoter. We transduced human iPSC-derived cardiomyocytes and engineered heart tissues (EHTs) carrying a compound heterozygous truncatingMYBPC3mutation (MYBPC3-/-) with BMN 293 and assessed cMyBP-C levels and contractile parameters. We also systemically administered BMN 293 to MYBPC3-/-mice and assessed cardiac distribution of human cMyBP-C by molecular and histological methods and determined its impact on left ventricular hypertrophy and function by echocardiography and other imaging techniques.Results:BMN 293 transduction of human iPSC MYBPC3-/-cardiomyocytes and EHTs resulted in high levels of humanMYBPC3mRNA and cMyBP-C protein, correct incorporation of cMyBP-C into the sarcomere, and complete normalization of contractile kinetics. BMN 293 was well tolerated in MYBPC3-/-mice and resulted in uniform restoration of cMyBP-C expression throughout the heart and significant correction of structural and functional cardiac abnormalities.Conclusions:A single IV infusion of BMN 293 to MYBPC3-/-mice resulted in early and sustained reduction in left ventricular hypertrophy and durable improvements in diastolic function.
Circulation, Volume 148, Issue Suppl_1, Page A18831-A18831, November 6, 2023. Introduction:Epidemiologic studies indicate that Blacks are protected against abdominal aortic aneurysm (AAA), despite similar risk factors as Caucasians. Approximately 70% of Blacks harbor a variant in the Duffy Antigen Receptor for Chemokines (DARC), a non-signaling receptor expressed primarily on erythrocytes which binds and regulates inflammatory chemokines. Since chemokines play a critical role in AAA, we tested the hypothesis that lack of DARC expression on erythrocytes protects against AAA, and we investigated the mechanisms.Methods:Mice lacking DARC on hematopoietic cells were created by mating DARC flox/flox mice with Vav1-Cre mice. At 10-14 weeks of age, both Vav1-Cre positive (DARCHKO) and Vav1-Cre – mice [wild type (WT) littermate controls] received PCSK9 adeno-associated virus injection and were started on high fat diet. After 1 week, mice were infused with angiotensin II (AngII, 1,000 ng/kg/min) via osmotic minipump for 26 days to induce AAA.Results:Compared to WT, there was no difference in blood pressure or cholesterol levels in AngII-infused DARCHKOmice, which were confirmed to lack DARC on erythrocytes by flow cytometry. Aortic diameter and AAA incidence were significantly decreased in DARCHKOcompared to WT mice (n=16-20, p=0.0257), in conjunction with reduced elastin degradation and macrophage infiltration. We did not find differences in levels of DARC-bound chemokines that could account for the protection against AAA. Interestingly, we found that the level and activity of lysyl oxidase (LOX), a cytokine-like enzyme that crosslinks extracellular matrix proteins and protects against AAA, were higher in DARCHKOmice compared to WT mice.Conclusions:Loss of DARC on hematopoietic cells protects against AAA formation, likely by upregulating aortic LOX. These findings provide novel insight into ethnic differences in susceptibility to AAA.
Circulation, Volume 148, Issue Suppl_1, Page A18729-A18729, November 6, 2023. BACKGROUND:Heart failure (HF) due to ischemic dilated cardiomyopathy (IDCM) is a major cause of morbidity and mortality. In failing hearts, loss of T-tubule microdomains is central to impaired calcium transients and loss of cardiac function. Cardiac bridging integrator 1 (cBIN1), a membrane scaffolding protein that organizes T-tubule microdomains, is transcriptionally reduced in failing hearts.Hypothesis:cBIN1 gene therapy will improve cardiac function in ischemic cardiomyopathy.AIM:Evaluate the effectiveness of catheter-based delivery of cBIN1 gene therapy in preserving bipolar voltage (an assay of muscle health) and LVEF in a canine model of IDCM.Methods:Nine healthy adult mongrel dogs (30±3 kg) underwent LAD ligation, developing IDCM and HFrEF (LVEF < 40% and NT-proBNP >900 pmol/L) over 3-4 months. Animals received a single treatment at 20 sites by intramyocardial injection of either AAV9-GFP (control, n=3) or AAV9-cBIN1 (n=6) using a MyoStar catheter-mediated subendocardial injection. Low voltage LV regions were quantified as percent of total endocardial area with bipolar voltage
Circulation, Volume 148, Issue Suppl_1, Page A16199-A16199, November 6, 2023. Introduction:Postsurgical Takotsubo cardiomyopathy (TCM) is associated with acute stress secondary to catecholamine release causing reversible left ventricular (LV) failure. We present a diagnostically challenging presentation of postsurgical TCM with underlying prothrombin gene mutation.Case Presentation:A 53 year old female underwent laparoscopic hysterectomy 2 days prior to presentation with chest pain and shortness of breath. Vitals were unremarkable, labs showed elevated troponin and EKG consistent with NSTEMI. CTA chest demonstrated significant clot burden to the ascending and descending aorta. Echocardiogram revealed a reduced ejection fraction (EF) of 25-30% with apical ballooning and thrombus. She was admitted on heparin drip and completed an infusion of eptifibatide for 24 hours. Repeat CTA chest demonstrated mildly reduced clot burden. Left heart catheterization (LHC) was precluded due to aortic arch clot. Coronary CTA revealed no CAD with a calcium score of 0. Coagulopathy workup showed heterozygous prothrombin G20210A variant. She was discharged on warfarin. Repeat echocardiography 1 month later showed improved EF to 40-45%.Discussion:TCM accounts for 1-3% of cases of acute coronary syndromes (ACS) and affects women at a rate of 90%. Studies have shown that TCM with apical ballooning is minimally associated with LV thrombus . The presence of heterozygous prothrombin gene mutation increases risk of clot formation by 2 to 3 times. Surgery can cause this risk to dramatically increase. Coronary CTA was the best noninvasive approach to make the diagnosis as the risk of LHC was too great given the anatomic challenge of the aortic arch clot. We advocate for hypercoagulable workups to inform management of anticoagulation in patients with clotting events without known risk factors for thrombus formation.
Circulation, Volume 148, Issue Suppl_1, Page A18022-A18022, November 6, 2023. Hypertrophic cardiomyopathy (HCM) is an inheritable cardiac condition affecting 1:200-500 people and is characterized by left ventricular hypertrophy that can cause heart failure. Autosomal-dominant, missense mutations inMYH7(encoding MHC-β) cause 33% of genotype+HCM cases. Despite this, 75% ofMYH7missense variants are of unknown clinical significance (VUS). Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can elucidate the effect(s) of VUS, however, new variant identification by genetic testing outpaces methods to gene edit hiPSCs. Thus, we developed a method calledCRISPRaOn-TargetEditingRetrieval (CRaTER) which increased successful gene-editing 25-fold over current methods. We leveraged CRaTER to enrich for edited hiPSCs, generating a library of 113MYH7missense variants. As mutations can reduce protein stability and abundance, we flow sortedMYH7variant hiPSC-CMs into bins based on differing MHC-β protein abundance followed by next generation sequencing to calculate variant abundance scores. This multiplexed assay identified 31 residue substitutions as functionally abnormal, including all tested pathogenic variants and VUS. MHC-β depletion was validated in clonal hiPSC-CMs and by Western blotting of patient myocardium with aMYH7mutation. Additionally, we found that severe MHC-β depletion is associated with increased cell size, a hallmark pathological phenotype of HCM. Finally, single-cell contractility analyses of hiPSC-CMs with a pathogenicMYH7variant revealed hyperdynamic physiology relative to isogenic controls, suggesting an association between MHC-β loss and systolic dysfunction. Together, CRaTER enables the generation of isogenic hiPSC variant libraries at unprecedented scale for differentiation to disease-relevant cell types. Functional analyses ofMYH7variant hiPSC-CMs have revealed a novel correlation between MHC-β protein depletion and pathogenicity that accurately predictsMYH7variant effect and has implications for futureMYH7variant effect interpretations.
Circulation, Volume 148, Issue Suppl_1, Page A14717-A14717, November 6, 2023. Heart failure (HF) is a major cause of morbidity and mortality worldwide, yet with limited therapeutic options. Recent evidence indicates that the cardiac bridging integrator 1 (cBIN1)-organizes transverse tubule microdomains which organize the calcium handling machinery important for cardiac function. In rodent models, adeno-associated virus 9 (AAV9)-cBIN1 effectively reconstitutes microdomains and rescues HF. Here we explored the therapeutic efficacy of AAV9-cBIN1 in a minipig model of non-ischemic HF. 6-7 months old Yucatan minipigs were subjected to continuous right ventricular rapid pacing (RVP) at 170 bpm. After continuous RVP for 5.7 ± 1.0 weeks (N=10), minipigs developed dilated cardiomyopathy with increased left ventricular (LV) end diastolic volume (EDV, 70.7 ± 4.5 vs. baseline 45.5 ± 1.6 mL) and reduced ejection fraction (EF, 36.5 ± 1.2 vs. baseline 71.3 ± 1.5 %). Once EF ≤ 40% over a week, pigs were randomized to receive intravenous AAV9-cBIN1 or control (AAV9-GFP or PBS). Pacing was continued throughout the study period. In control animals (N=5 with 4 PBS and 1 AAV9-GFP treatment), LV further dilated (78.0 ± 8.7 mL) with EF declining to 27.6 ± 6.7 % by four weeks post injection. The first three control animals already died of severe HF symptoms by 7 weeks post injection (Figure 1A) and the other two animals are currently at 4 weeks post injection with EF reduced to 27 and 33 %, respectively. Despite of continuous RVP, AAV9-cBIN1 rescued HF with EF recovered to 52.6 ± 4.0 % and EDV at 67.3 ± 6.6 mL at 4 weeks post injection. Three cBIN1-treated animals survived the full protocol of 6-month monitoring with EF maintained at 57.5 ± 15.5 % (Figure 1B-C), with two ongoing pigs currently surviving at 4 weeks post-injection with normal EF. Figure contains data of the animals who died or have already completed the 6-month protocol (N=3 per group). No toxicity was observed. In conclusion, intravenous cBIN1 gene therapy rescues non-ischemic HF and improves mortality in pigs.
Circulation, Volume 148, Issue Suppl_1, Page A18721-A18721, November 6, 2023. Introduction:With a prevalence of up to 1 in 200, hypertrophic cardiomyopathy (HCM) is the most common monogenic hereditary cardiovascular disease. Previous studies have observed that mitochondrial dysfunction and mitochondria-associated metabolic remodeling are major metabolic features in HCM. However, the impact of mitochondrial gene mutations on HCM were poorly understood.Methods and Results:We selected 2272 mitochondrial genes from the Mitocarta database 3.0 and screened rare variants from the whole-exome sequencing data in 985 HCM patients (excluding patients with HCM phenotypes, including mitochondrial myopathy) and 759 non-HCM controls. We found thatCYP2E1rare variants were significantly enriched in HCM patients (OR = 9.56, Bonferroni corrected P = 0.001). Patients with or withoutCYP2E1rare variants shared similar demographic and echocardiographic variables at baseline. With a median follow-up time of 5.31 years in 735 HCM patients, we revealed that HCM patients withCYP2E1rare variants were more likely to experience myocardial infarction (HR 14.1, P < 0.001).Conclusion:We demonstrated that rare variants in the mitochondrial geneCYP2E1might increase the risk of HCM onset and may be associated with the occurrence of myocardial infarction endpoints in HCM. We hypothesize that the mechanism is related to the extensive and complex mitochondrial metabolic processes in whichCYP2E1is involved. Our findings provided new evidence for the association between mitochondrial gene variants and HCM, and more multi-center evidence is needed to confirm this association in the future.
Circulation, Volume 148, Issue Suppl_1, Page A12016-A12016, November 6, 2023. Introduction:Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disease and may lead to tragic outcomes such as sudden cardiac death and heart failure. Previous studies have shown that HCM patients undergo significant lipid-related metabolic reprogramming. But whether lipid gene variants affect the diagnosis and prognosis of HCM patients remains unclear.Methods and Results:Whole-exome sequencing was performed in 985 patients with HCM and 759 non-HCM controls to search for lipid gene variants. TheAPOErare variants were more prevalent in patients with HCM than controls (odds ratio, 4.91, P < 0.001). Patients withAPOErare variants have higher left ventricular outflow tract gradient (LVOTG) (71.0 VS 38.0 mmHg, P = 0.035), higher triglycerides (1.59 VS 1.32 mmol/L, P = 0.051) and lower lipoprotein a (65.5 vs 113 mg/L, P = 0.076) in peripheral blood.APOEcarriers also had elevated free fatty acids in 290 surgically resected myocardial tissue lipidomics data (P = 0.008). With a median follow-up time of 5.10 years, HCM patients withAPOErare variants were more likely to experience cardiovascular death (adjusted HR 4.8, P = 0.001) and all-cause death (adjusted HR 3.5, P = 0.008).Conclusion:Our cohort data indicated that rare variants ofAPOEare associated with altered lipid metabolism in peripheral blood as well as myocardial tissue, increased LVOTG, and greater risk of HCM disease-onset as well as poor prognosis. We proposed thatAPOEmight be associated with HCM and that its contribution to myocardial hypertrophy through lipotoxicity may be an emerging mechanism for HCM pathogenesis and progression.
Circulation, Volume 148, Issue Suppl_1, Page A12986-A12986, November 6, 2023. Introduction:Thoracic aortic disease is an important cause of morbidity and mortality in the US, and aortic diameter is a heritable contributor to risk.Question:Could a polygenic prediction of ascending aortic diameter improve detection of aortic aneurysm?Methods:We used deep learning to measure ascending thoracic aortic diameter in 56,556 UK Biobank participants and conducted a genome-wide association study (GWAS) in 39,524. We built a 1.1-million SNP polygenic score usingPRScs. In 4,896 non-GWAS participants (“training set”), we built linear models from clinical variables and the polygenic score (“AORTA Gene”) and simpler comparators. The models were assessed in the remaining 4,962 unrelated UK Biobank participants with imaging (“test set”). The models were externally validated inAll Of Usparticipants over the age of 40.Results:In the test set, the AORTA Gene model explained 39.9% (95% CI 37.8-42.0%) of the variance in thoracic aortic diameter compared to 29.2% for the clinical model (95% CI 27.1-31.4%). For diameter ≥ 4cm, the area under the receiver operating characteristic curve (AUROC) was 0.834 for AORTA Gene, superior to the clinical model (0.765, P=7.3E-10). Thresholding at the top 10% of the score, AORTA Gene had a sensitivity of 52% and a specificity of 91.2% (F1: 0.197) for detecting the 113 participants with diameter ≥ 4. For the clinical score, the respective values were 36.3%, 90.9%, and 0.137.In 610All Of Usparticipants with genetic data and aortic measurements, the AORTA Gene model had a correlation of 0.591 (95% CI 0.537-0.640) with aortic diametervs0.538 (95% CI 0.479-0.592) for the clinical model (P=2.2E-13 against a null hypothesis of the genetics being uninformative beyond clinical factors). It had an AUROC of 0.827 for identifying diameter ≥4cmvs0.791 for the clinical model (P=7.8E-03). AORTA Gene also had a higher AUROC for prevalent thoracic aneurysm diagnoses (0.760vs0.739 for the clinical score, N=1,904 cases, P=2.4E-16) and incident diagnoses (0.748vs0.729, N=1,632 events, P=9.5E-10).Conclusions:Genetic information improved prediction of thoracic aortic diameter and aneurysm when added to clinical risk factors. Larger and more diverse samples will be needed to develop more powerful scores.
Circulation, Volume 148, Issue Suppl_1, Page A13924-A13924, November 6, 2023. Introduction:Atrial fibrillation (AF) is a multifactorial disease, with oxidative stress (NADPH oxidase 2, NOX2) and autonomic remodeling (hyperinnervation and parasympathetic signaling) playing an important role. Disrupting both mechanisms simultaneously with gene-based therapy may allow for reversal of established remodeling in AF.Hypothesis:Gene therapy aimed at attenuation of parasympathetic signaling (Gαi/o inhibitory peptides, GiGo_ct) and prevention of nerve sprouting (nerve growth factor short hairpin RNA, NGF shRNA) combined with reduction of oxidative stress (NOX2 shRNA) can reverse AF in a large animal chronic AF model.Methods:Chronic AF model was created by rapid atrial pacing (RAP) at 600bpm in 5 dogs, followed by gene therapy treatment (Panel A). A combination of NOX2 shRNA and GiGo_ct plasmids with (n=2) or without (n=3) NGF shRNA was injected in both atria followed by electroporation to facilitate gene delivery. Control RAP dogs did not receive gene (n=5) or underwent scrambled gene injection (n=6). AF burden was assessed over time and residual AF was recorded at terminal EP study. NOX2 and NGF knockdown and GiGo_ct expression were evaluated by PCR.Results:After 3.3±1.3 weeks of RAP, all 11 animals reached >80% AF burden. Combined (NOX2+GiGo±NGF) gene therapy significantly reduced AF burden by 66% (p
Circulation, Volume 148, Issue Suppl_1, Page A11668-A11668, November 6, 2023. Introduction:In aortic stenosis (AS), women present less aortic valve calcification and more aortic valve fibrosis than men for the same hemodynamic severity. However, the specific mediators that drive the fibro-calcific differences between men and women remain unclear. We aim to assess the transcriptome of stenotic aortic valves explanted during aortic valve replacement, according to patient’s sex.Methods:Transcriptomic profile was obtained from 240 explanted human aortic valves. Among these 240 patients (120 women and 120 men), 62 women were matched with 62 men for age (within 2 years), body mass index (within 2 kg/m2), arterial pressure (within 10/5 mmHg), diabetes (exact), hypertension (exact) and AS severity (Table 1). Genes were classified in 6 key processes of AS development: oxidative stress, inflammation, lipid metabolism, fibrosis, apoptosis and calcification following a literature review.Results:One hundred and ninety (190) genes were differently regulated between men and women: 132 on autosomes and 58 on sexual chromosomes. Among these genes, 106 were over-expressed and 84 were under-expressed in women compared to men (Figure 1). Different genes involved in processes of inflammation, lipid metabolism and calcification were up-regulated both in women and men (women:NET1,KIF1A, CES1, RCN2;men:FERMT3, APOD, CPAMD8, STC2). Genes involved in apoptosis (SFRP4) and fibrosis processes (TGFβ2,FRAS1) were overexpressed in women.Conclusions:This study provides evidence that sex may influence aortic valve gene expression through different mechanisms in females and males, favoring pro-fibrotic and pro-apoptotic processes in women.
Circulation, Volume 148, Issue Suppl_1, Page A13953-A13953, November 6, 2023. Background:Genome-wide association studies (GWAS) have improved our understanding of the genetic basis of coronary artery disease (CAD). While sex-differences in CAD have been observed, little is known about the role of sex hormones in CAD genetics. Accounting for sex hormone levels may help identify CAD-risk loci and extend our knowledge of its genetic architecture.Methods:We performed a GWAS to identify CAD risk loci by incorporating gene-sex hormone interactions in 365,662 individuals of European ancestry enrolled in the UK Biobank (UKB). Genetic interactions of total testosterone (TT), bioavailable testosterone (BAT) and sex-hormone binding globulin (SHBG) were evaluated. GEM software was used to conduct sex-stratified genome-wide interaction analysis with prevalent CAD. Enrollment age and principal components 1-10 were model covariates. The online post-GWAS tool FUMA was used to annotate the biological functions of marginal, interaction, and joint effects summary statistics.Results:We identified 64 loci in males and 8 loci in females that reached genome-wide significance (p < 5х10-8) for CAD (Table 1). Among males, 3 significant loci were identified for BAT, one for TT and one for SHBG using joint effect model that did not overlap with those identified with marginal effect. Among females, 5 significant loci were identified, all for TT using joint effect that that did not overlap with those identified with marginal effect. Of these 5 loci, two loci were independently identified with significant SNP-TT interaction. These loci were mapped close to the TRDN and THADA genes that have been implicated in congenital ventricular tachycardia and polycystic ovarian syndrome, respectively.Conclusion:This Genome-wide gene-sex hormone interaction study identified genomic-risk loci that may contribute to the differential CAD risk between men and women, which would otherwise not have been discovered in traditional GWAS only including marginal genetic effect.
Circulation, Volume 148, Issue Suppl_1, Page A14481-A14481, November 6, 2023. Background:Adeno-associated virus (AAV) based gene therapy bears the potential to transform future clinical care of hereditary and acquired heart failure. However, the broadly used AAV9 serotype exhibits fatal liver toxicity in clinical studies and requires long-term immunosuppressive therapy when used systemically e.g., for LAMP2 gene-associated cardiomyopathy.Hypothesis:AAVs with genetically-engineered cardiac tropism are key for safe and efficient delivery of therapeutic gene replacement, silencing or editing systems to the diseased heart with minimized off-target organ transduction after a simple systemic administration.Technology & Results:We developed novel cardiac-specific AAV capsids by a next-generation experimental-bioinformatic AAV capsid development platform (next-CAP) employing mouse, farm pig and non-human primate models. A highly diverse capsid library with more than 100 million variants was generated from novel mammalian heart AAV isolates integrating DNA shuffling and peptide display approaches and underwent a two-step cross-speciesin vivoevolution. Implementing a unique molecular identifier strategy eventually allowed for combined long- and short-term sequencing to capture 77 novel AAV capsid variants displaying highest enrichment in hearts over all other organs, including the liver with superior patterns to AAV9. Subgroups of the top heart-AAV capsid portfolio displayed an overall sequence homology of up to 99.5% identity highlighting the presence of novel cross-species conserved heart-homing motifs. As producibility was another inherent screening criteria, our cardiac-specific AAV capsid portfolio already provides scalable manufacturing characteristics for in vivo indications.Conclusion:Our proprietary portfolio of novel heart-specific AAV capsids now enables the systematic development of optimal therapeutic capsid-promoter-transgene ensembles for further dose-expression, -efficacy and -toxicity assessments for various hereditary and acquired HF indications in suitable model systems. In summary, we developed an efficient and versatile platform for the generation of next-generation AAV capsids for heart-specific gene therapy of yet uncurable rare and common cardiac diseases.