New Years Schedule Update

As we celebrate the start of the New Year, our customer service team will be unavailable after 12pm EST December 31st, and will return on Friday, January 2nd.
Due to increased seasonal volume, fulfillment times may be slightly extended.
We appreciate your patience and wish you Happy New Year!

PLEASE LOGIN TO SEE OUR RESEARCH PEPTIDES
Click Here
US MANUFACTURED RESEARCH USE ONLY PEPTIDES
Click Here

SS-31: Targeting Mitochondrial Dysfunction

SS-31 10mg

SS-31: A Case Study in Targeting Mitochondrial Dysfunction

I. The Mitochondrion as a Therapeutic Frontier

1.1 Beyond the Powerhouse: The Centrality of Mitochondria in Cellular Health

The mitochondrion, long simplified in biological vernacular as the cellular “powerhouse,” is now understood to be a dynamic and pleiotropic organelle that functions as a central signaling and metabolic hub, indispensable for eukaryotic life. Its integrity and function are paramount not only for energy production but also for a vast array of processes that dictate cellular fate and maintain organismal homeostasis. A sophisticated appreciation of its multifaceted roles is essential to contextualize the profound consequences of its dysfunction and to recognize its emergence as a compelling therapeutic frontier.

Core Function – Oxidative Phosphorylation (OXPHOS)

The canonical role of the mitochondrion is the synthesis of adenosine triphosphate (ATP), the universal energy currency of the cell, through the process of oxidative phosphorylation (OXPHOS).1 This highly efficient metabolic pathway is the culmination of cellular respiration, converting the chemical energy stored in nutrients into a usable form. The process begins upstream with catabolic pathways such as glycolysis and fatty acid beta-oxidation, which break down glucose and fats to produce key intermediates. These intermediates feed into the citric acid cycle (or Krebs cycle) within the mitochondrial matrix, generating high-energy electron carriers, primarily nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FADH2).2

These electron carriers are the essential fuel for the electron transport chain (ETC), a series of five large protein-lipid enzyme complexes (designated Complexes I-V) embedded within the highly folded inner mitochondrial membrane (IMM).2 NADH and FADH2 donate their electrons to the ETC at Complex I and Complex II, respectively. The electrons are then passed sequentially through the chain—from Complex I/II to coenzyme Q10, then to Complex III, cytochrome c, and finally to Complex IV—in a series of precisely controlled oxidation-reduction reactions.3 The ultimate electron acceptor at the end of this chain is molecular oxygen, which is reduced to form water.3

The transfer of electrons is a highly exergonic process, and the energy released at Complexes I, III, and IV is harnessed to actively pump protons () from the mitochondrial matrix across the IMM into the intermembrane space.3 This action establishes a powerful electrochemical gradient, comprising both a pH differential (the matrix becomes alkaline, around pH 8) and a voltage differential (the matrix becomes negative, approximately -140 mV).4 This stored potential energy is termed the ‘proton motive force’.3 The final stage of OXPHOS is catalyzed by Complex V, more commonly known as ATP synthase. This remarkable molecular machine acts as a rotary motor; as protons flow back down their electrochemical gradient into the matrix through a channel in the  subunit of ATP synthase, they drive the rotation of the  subunit, which in turn catalyzes the phosphorylation of adenosine diphosphate (ADP) to ATP.4 The efficiency of this system is staggering: the complete oxidation of a single glucose molecule via OXPHOS yields approximately 30 to 32 molecules of ATP, a dramatic increase over the mere two ATP molecules produced by glycolysis alone.3 This immense energy output is what fuels virtually all essential biological functions.

Integral Hub for Cellular Homeostasis

Beyond its bioenergetic role, the mitochondrion is a critical nexus for numerous other cellular processes. It is the gatekeeper of the intrinsic pathway of apoptosis, or programmed cell death.5 This process is fundamental for embryonic development (e.g., sculpting digits), tissue homeostasis (balancing cell division with cell death), and the elimination of damaged or potentially cancerous cells.5 The release of mitochondrial intermembrane space proteins, most notably cytochrome c, into the cytosol is a key initiating event that triggers a cascade of caspase proteases, leading to the orderly dismantling and disposal of the cell.5

Mitochondria are also central to maintaining cellular calcium homeostasis, sequestering and releasing calcium ions to modulate a wide range of signaling pathways. Furthermore, as a natural consequence of electron transport, mitochondria are the primary intracellular source of reactive oxygen species (ROS)—chemically reactive molecules like superoxide radicals that are formed when electrons prematurely leak from the ETC and react with oxygen.8 While ROS play roles in cell signaling at low levels, their overproduction leads to oxidative stress, a damaging state where cellular components like lipids, proteins, and DNA are oxidized, contributing to cellular dysfunction and aging.8 The mitochondrion is thus a finely balanced organelle, where efficient energy production is inextricably linked to the control of cell death and redox signaling.

1.2 Mitochondrial Dysfunction as a Unifying Pathogenic Hub

Given the organelle’s centrality, it is logical that its failure, termed “mitochondrial dysfunction,” is a profoundly pathogenic state. This term encompasses a spectrum of defects, including impaired ATP production, leading to a cellular energy crisis; excessive generation of ROS, causing widespread oxidative damage; dysregulated calcium buffering, which disrupts cellular signaling; and the inappropriate activation of apoptotic pathways, leading to unwarranted cell death.

This pathological state has been identified as a core, unifying component in a vast and diverse array of human diseases. These range from rare, monogenic mitochondrial diseases, such as Barth syndrome and primary mitochondrial myopathies, to highly prevalent chronic conditions.9 The consequences of mitochondrial failure are most pronounced in tissues and organs with high metabolic demands and a heavy reliance on OXPHOS for their function. The heart, which produces and consumes approximately 6 kg of ATP daily, is exquisitely vulnerable, making mitochondrial dysfunction a key driver of cardiomyopathy and heart failure.11 The brain and central nervous system are similarly susceptible, with mitochondrial impairment implicated in the pathogenesis of neurodegenerative diseases like Parkinson’s and Alzheimer’s disease. Skeletal muscle, which requires immense energy for contraction, is the primary site of pathology in mitochondrial myopathies, characterized by weakness and exercise intolerance.10 The photoreceptors of the retina, which have one of the highest metabolic rates in the body, are also highly vulnerable, linking mitochondrial dysfunction to the progression of diseases like dry age-related macular degeneration (AMD).14 This growing understanding of mitochondrial dysfunction as a common pathogenic denominator across numerous debilitating conditions has firmly established the mitochondrion as one of the most attractive and promising targets for novel therapeutic intervention.

II. The Mechanism of Action: A Masterclass in Molecular Targeting

The therapeutic promise of SS-31 is rooted in its elegant and highly specific mechanism of action, which targets the fundamental underpinnings of mitochondrial dysfunction. The scientific narrative of this mechanism has evolved significantly, moving from an initial characterization of the compound as a simple antioxidant to a more sophisticated understanding of it as a fundamental stabilizer of the bioenergetic machinery of the inner mitochondrial membrane. This refined view explains its profound and pleiotropic restorative effects on cellular health.

2.2 The Role of Supercomplexes in Optimizing Electron Transport

For decades, the “fluid mosaic” model depicted the ETC complexes as discrete, freely diffusing entities within the IMM. However, modern biochemical research has overturned this view, revealing that these complexes assemble into large, functionally active, supramolecular structures known as “respiratory supercomplexes” or “respirasomes”.16 The most common of these assemblies brings together Complexes I, III, and IV into a single, highly efficient unit.16

The formation of these supercomplexes is not merely an incidental aggregation; it confers significant functional advantages. By physically associating the components of the ETC, supercomplexes are thought to enhance the efficiency of electron transfer through a process known as substrate channeling, minimizing the distance that mobile electron carriers like coenzyme Q10 and cytochrome c must travel.17 This optimized organization enhances the overall catalytic activity of the respiratory chain.16 Crucially, this tight coupling also reduces the premature leakage of electrons from the chain, thereby minimizing the production of damaging ROS.18 The stability and assembly of these vital supercomplexes are heavily dependent on the lipid composition of the IMM, and in particular, require the presence of cardiolipin, which acts as a “molecular glue” to hold the structures together.16 Consequently, the oxidative damage to cardiolipin seen in pathological states leads to the dissociation of these supercomplexes, resulting in a less efficient, “leaky” ETC that produces less ATP and more ROS—a core feature of mitochondrial dysfunction.

2.3 SS-31’s Biochemical Profile and Targeted Interaction with Cardiolipin

SS-31 (also known by the developmental codes SS-31 and MTP-131) is a synthetic, water-soluble tetrapeptide.19 Its specific amino acid sequence is D-Arginyl-2′,6′-dimethyl-L-tyrosyl-L-lysyl-L-phenylalaninamide (H-D-Arg-Dmt-Lys-Phe-NH2).20 The molecule’s architecture is the key to its function, featuring an alternating motif of aromatic residues (dimethyltyrosine, phenylalanine) and basic amino acids (arginine, lysine). At physiological pH, the arginine and lysine residues are protonated, conferring a net positive charge of +3 to the peptide, making it strongly cationic.20

This positive charge is the critical determinant of its pharmacological behavior. SS-31 is able to readily penetrate cell membranes in an energy-independent and non-saturable manner, suggesting it does not rely on specific transporters or receptors.19 Once inside the cell, its cationic nature drives a powerful electrostatic attraction to the highly negative membrane potential of the IMM, which is typically around -140 mV to -180 mV.4 This attraction causes the peptide to selectively target and accumulate at the IMM. Studies have demonstrated that SS-31 can be concentrated by up to 5000-fold within this subcellular compartment compared to the surrounding cytoplasm.19 Its primary binding partner at the IMM is the abundant, negatively charged phospholipid, cardiolipin.12 The strong electrostatic interaction between the peptide’s three positive charges and the two negative phosphate head groups of cardiolipin molecules anchors SS-31 to the membrane, where it can exert its therapeutic effects.21

PropertyDescription
Chemical Name / CodesElamipretide; SS-31; MTP-131; Bendavia; FORZINITY™
StructureTetrapeptide: D-Arginyl-2′,6′-dimethyl-L-tyrosyl-L-lysyl-L-phenylalaninamide
Key FeaturesAromatic-cationic motif; Net charge of +3 at physiological pH
Molecular TargetCardiolipin (CL) at the Inner Mitochondrial Membrane (IMM)
Cellular UptakePassive, energy-independent, non-saturable
Subcellular AccumulationUp to 5000-fold concentration at the IMM, driven by membrane potential
AdministrationDaily subcutaneous (SC) injection (in late-stage trials)
ExcretionRenal; nearly 100% of administered dose recovered in urine within 24 hours

2.4 From Molecular Binding to Cellular Restoration: Re-coupling the Engine of the Cell

The binding of SS-31 to cardiolipin initiates a cascade of restorative events that address the core deficits of mitochondrial dysfunction at their source. This mechanism is multifaceted, involving structural, bioenergetic, and anti-apoptotic effects.

Restoration of Cristae Architecture and ETC Supercomplex Stability

By associating with cardiolipin, SS-31 is theorized to shield this critical phospholipid from the damaging effects of oxidative stress.12 This protective interaction helps to preserve the structural and functional integrity of the IMM. A foundational effect is the restoration of proper cristae architecture, which is often disorganized and fragmented in diseased states.20 Re-establishing the correct membrane curvature and folding is a prerequisite for efficient energy production. Concurrently, the SS-31-cardiolipin interaction promotes the stability and re-assembly of the ETC supercomplexes.22 In states of mitochondrial dysfunction where oxidative damage to cardiolipin has caused these supercomplexes to dissociate, SS-31 helps to restore their crucial organization. This structural stabilization is the first and most fundamental step in its restorative action.

Enhancement of ATP Synthesis

The direct and most important consequence of a more organized membrane and a more stably coupled ETC is a significant improvement in mitochondrial respiration and ATP synthesis.19 By promoting the proper function of the ETC supercomplexes, SS-31 ensures a more fluid and efficient transfer of electrons along the respiratory chain, which in turn enhances the process of oxidative phosphorylation.20 This leads to a restoration of cellular ATP levels, providing energy-starved cells with the fuel required for normal function, maintenance, and repair.19 Further research has elucidated an even more direct role in this process. SS-31 has been shown to bind directly to key components of the ATP transport and production machinery, including the Adenine Nucleotide Translocator (ANT) and ATP Synthase (Complex V) itself.24 This interaction appears to improve the mitochondrion’s sensitivity to ADP, the primary substrate for ATP synthesis, thereby increasing the overall rate and efficiency of ATP production.24

Dual-Mechanism Mitigation of Oxidative Stress

SS-31 mitigates the damaging effects of oxidative stress through two distinct and complementary mechanisms. Initial hypotheses regarding its function centered on its direct antioxidant properties. However, a more comprehensive understanding has emerged, repositioning its primary action further upstream, at the very source of pathological ROS production.

  1. Upstream Prevention (Primary Mechanism): The principal source of pathological ROS in the cell is a “leaky” and inefficient electron transport chain. When supercomplexes are disorganized, electron transfer is impaired, allowing electrons to escape the chain prematurely and react with molecular oxygen to form superoxide radicals.18 By stabilizing cardiolipin and improving the coupling and efficiency of the ETC, SS-31 fundamentally reduces this electron leakage.19 This action prevents the formation of ROS at its source and is now considered the principal mechanism by which SS-31 exerts its profound antioxidant effects. This upstream prevention is a far more powerful and fundamental therapeutic action than mere downstream scavenging.
  2. Downstream Scavenging (Secondary Mechanism): The inclusion of a dimethyltyrosine (Dmt) residue in SS-31’s structure endows it with the ability to act as a direct free radical scavenger.19 This modified amino acid can directly interact with and neutralize existing oxygen radicals, such as hydroxyl and peroxynitrite radicals, thereby inhibiting downstream damage like lipid peroxidation. This direct scavenging capacity serves as a secondary, supportive mechanism to the primary effect of preventing ROS formation.

This evolution in understanding—from viewing SS-31 as a simple ROS scavenger to recognizing it as a fundamental bioenergetic stabilizer—is critical. It explains why the compound has such profound effects on restoring ATP production and why it holds broad therapeutic potential across diseases characterized by a primary energy failure, a pathology that is more fundamental than oxidative stress alone.

Inhibition of Apoptosis

SS-31 also exerts potent anti-apoptotic effects by stabilizing the IMM. In healthy mitochondria, cardiolipin anchors cytochrome c to the inner membrane. During cellular stress, oxidative damage can cause cardiolipin to translocate to the outer mitochondrial membrane, facilitating the release of cytochrome c into the cytosol—a critical, committing step in the intrinsic apoptotic cascade.5 By binding to and stabilizing cardiolipin, SS-31 helps to prevent this release, thereby inhibiting the initiation of programmed cell death.12 It has also been shown to inhibit the opening of the mitochondrial permeability transition pore (mPTP), a large, non-selective channel whose prolonged opening leads to mitochondrial swelling, rupture, and subsequent cell death.

A key theme that emerges from this mechanistic analysis is the compound’s remarkable selectivity for pathological states. Numerous studies, including ex vivo experiments on cardiac tissue from explanted failing human hearts, have demonstrated that SS-31 significantly improves function in diseased, energy-depleted mitochondria but has little to no effect on the function of healthy mitochondria.19 This suggests that SS-31 is not an indiscriminate metabolic “booster” but rather a targeted “stabilizer.” In a healthy mitochondrion, where cardiolipin is intact and the ETC machinery is already optimally configured into supercomplexes, there is no disorganized pathological substrate for the drug to act upon. Its effect is conditional on the presence of dysfunction. This selective action likely underpins its highly favorable safety profile and suggests a high therapeutic index, as it leaves normal cellular processes unperturbed.

III. Clinical Development: A Journey of Precision, Persistence, and Precedent

The clinical development of SS-31 has been a long and complex journey, spanning multiple distinct therapeutic areas where mitochondrial dysfunction is a core element of the disease pathophysiology. The program has yielded a rich tapestry of results—including both successes and setbacks—that have provided critical learnings, progressively refining the understanding of the drug’s potential and illuminating the immense challenges of treating these intricate diseases. The story of SS-31’s translation from bench to bedside is a case study in the importance of precision medicine, endpoint innovation, and regulatory persistence.

Trial NameIndicationPhasePrimary Endpoint(s)Key Outcome & Current Status
TAZPOWERBarth Syndrome (BTHS)2/3Change in 6-Minute Walk Test (6MWT)Blinded crossover did not meet endpoints.25 Open-label extension showed significant improvements in 6MWT, muscle strength, and cardiac stroke volume.26 NDA approved by FDA via accelerated pathway.27
MMPOWER-3Primary Mitochondrial Myopathy (PMM)3Change in 6MWT; Change in PMMSA Total Fatigue ScoreDid not meet primary endpoints in the overall population.28 Subgroup analysis showed significant 6MWT improvement in patients with nuclear DNA (nDNA) mutations.23
NuPOWERPMM (nDNA mutations)3Change in 6MWTTrial designed based on MMPOWER-3 subgroup findings; fully enrolled and ongoing.23
ReCLAIM-2Dry Age-Related Macular Degeneration (AMD) with Geographic Atrophy (GA)2Change in Low-Luminance BCVA; Change in GA areaDid not meet primary endpoints.30 Showed a 43% reduction in the rate of photoreceptor loss (EZ attenuation), a key anatomical endpoint.30
ReNEW / ReGAINDry AMD3Rate of change in macular area of photoreceptor loss (EZ attenuation)Two pivotal trials initiated based on ReCLAIM-2 results and FDA guidance; currently enrolling.15
EMBRACE-STEMIHeart Failure (post-ST-Elevation Myocardial Infarction)2aMyocardial infarct sizeDid not reduce infarct size. Was associated with a reduced incidence of new-onset heart failure post-procedure.
Phase 2a RenalAtherosclerotic Renal Artery Stenosis2aRenal function, oxygenation, and blood flowAdjunctive SS-31 attenuated post-procedural hypoxia, increased renal blood flow, and improved estimated GFR.

3.1 Barth Syndrome: Validating the Mechanism in a Prototypical Disease of Cardiolipin

Pathophysiology

Barth syndrome (BTHS) represents a prototypical disease of cardiolipin deficiency, making it an ideal indication to test the therapeutic hypothesis of SS-31. BTHS is an ultra-rare, X-linked genetic disorder caused by mutations in the TAFAZZIN gene.11 This gene encodes the enzyme tafazzin, which is responsible for the final remodeling step in cardiolipin biosynthesis.22 A defect in this enzyme leads to a profound deficiency of mature, functional cardiolipin and a concomitant accumulation of an abnormal precursor, monolysocardiolipin (MLCL).11 This altered lipid profile severely compromises mitochondrial structure and function, leading to the characteristic clinical triad of cardiomyopathy, skeletal myopathy with debilitating fatigue and exercise intolerance, and chronic or intermittent neutropenia.11

The TAZPOWER Trial

The pivotal clinical study for SS-31 in this population was the Phase 2/3 TAZPOWER trial. The initial part of the study was a 28-week, randomized, double-blind, placebo-controlled crossover trial.26 This rigorous design, however, failed to demonstrate a statistically significant benefit for SS-31 on the primary endpoints, which included the 6-Minute Walk Test (6MWT).25 This outcome was initially a significant setback.

However, the subsequent open-label extension (OLE) phase of the trial, where all participants received daily subcutaneous SS-31, yielded dramatically different and highly promising results. Over long-term treatment, patients demonstrated clinically meaningful and statistically significant improvements across multiple domains. By week 168 of the OLE, patients showed a cumulative improvement in the 6MWT distance of 96.1 meters from their OLE baseline.36 They also exhibited significant increases in muscle strength as measured by handheld dynamometry and improved scores on patient-reported outcome measures for fatigue.21 Perhaps most notably, long-term treatment was associated with significant improvements in cardiac function. Echocardiographic analysis revealed a 27% increase in average cardiac stroke volume and favorable trends in left ventricular end-diastolic and end-systolic volumes, suggesting positive cardiac remodeling was occurring.12 These functional gains were correlated with favorable changes in the MLCL:CL biomarker ratio, providing a physiologic basis for the observed clinical benefits.35

The discrepancy between the short-term crossover results and the long-term OLE results points to a critical mismatch between the trial design and the biological reality of the disease. BTHS is a chronic condition characterized by long-standing structural and functional deficits in high-energy tissues. The mechanism of SS-31 involves stabilizing membranes and improving bioenergetics, which would theoretically enable a slow process of cellular repair and tissue remodeling. The TAZPOWER results strongly suggest that this restorative process takes significantly longer than the 12 weeks allotted in the crossover phase to manifest in measurable functional gains. A short-term trial design may be fundamentally unsuited to capture this type of therapeutic benefit.

The Regulatory Saga

The regulatory journey for SS-31 in BTHS has been a protracted and complex saga, reflecting the broader challenges of drug development for ultra-rare diseases. Following the mixed results of TAZPOWER, Stealth BioTherapeutics’ New Drug Application (NDA) initially received a “refusal to file” letter from the U.S. Food and Drug Administration (FDA) in 2021, which cited the lack of a single, adequate, well-controlled trial.37

Despite this, persistent and organized advocacy from the patient community, their families, and clinical experts ultimately led the FDA to convene a Cardiovascular and Renal Drugs Advisory Committee meeting in October 2024.38 This meeting resulted in a 10-6 vote concluding that the drug was effective, a non-binding but influential recommendation in favor of approval.39 Following further review and delays, the FDA issued a complete response letter in May 2025, declining to approve the application in its current form but, for the first time, proposing a viable path forward for resubmission for accelerated approval.37 This path was based on using the improvement in knee extensor muscle strength—which had increased by over 45% in the TAZPOWER trial—as an intermediate clinical endpoint that is reasonably likely to predict clinical benefit.39

Following a rapid resubmission by the company in August 2025, the FDA granted Accelerated Approval to FORZINITY™ (SS-31) on September 19, 2025, for the treatment of Barth syndrome in patients weighing at least 30 kg.27 This landmark decision represents the first-ever approved therapy for BTHS and the first approved mitochondria-targeted therapeutic. The approval journey is a powerful case study in modern drug regulation, demonstrating the FDA’s increasing willingness to exercise regulatory flexibility for diseases with high unmet need. It highlights the acceptance of evidence from OLEs, comparisons to natural history controls, and the use of novel intermediate endpoints when traditional, large placebo-controlled trials are unfeasible. It also underscores the transformative power of patient advocacy in shaping regulatory outcomes.

3.2 Primary Mitochondrial Myopathy: A Pivotal Lesson in Genetic Heterogeneity

Pathophysiology

Primary Mitochondrial Myopathies (PMM) are a clinically and genetically heterogeneous group of disorders characterized by impaired mitochondrial oxidative phosphorylation that predominantly affects skeletal muscle.10 The genetic basis can lie in either the mitochondrial DNA (mtDNA), which encodes 13 essential protein subunits of the ETC, or the thousands of genes in the nuclear DNA (nDNA) that are required for mitochondrial structure, function, and maintenance.10 Patients typically suffer from progressive muscle weakness, severe fatigue, and exercise intolerance.44

The MMPOWER-3 Trial

The MMPOWER-3 trial was a large, pivotal Phase 3 study designed to evaluate the efficacy of a 40 mg daily subcutaneous dose of SS-31 in 218 adults with genetically confirmed PMM.28 It was conceived as a “basket” study, enrolling a broad population of patients with a wide variety of underlying mtDNA and nDNA mutations. The trial ultimately failed to meet its co-primary endpoints: over 24 weeks, there was no statistically significant difference between the SS-31 and placebo groups in the change from baseline on either the 6MWT or the Primary Mitochondrial Myopathy Symptom Assessment (PMMSA) total fatigue score.28

The Crucial Subgroup Analysis

While the overall result was negative, the most significant finding from MMPOWER-3 emerged from a pre-specified subgroup analysis. This analysis revealed a striking divergence in treatment response based on the location of the pathogenic mutation. The subgroup of patients with nDNA mutations (n=59), particularly those with defects in genes required for mtDNA maintenance and replication (the “mtDNA replisome,” such as POLG and TWNK), demonstrated a statistically significant and clinically meaningful improvement in the 6MWT, walking on average 25.2 meters further than their placebo counterparts.23 Conversely, the larger subgroup of patients with primary mtDNA mutations (n=162) showed no improvement compared to placebo.23

This differential outcome provides a compelling, mechanistically plausible rationale for the trial’s overall result and represents a critical scientific learning. SS-31’s mechanism is to stabilize cardiolipin and organize structurally normal ETC proteins into functional supercomplexes. In patients with primary mtDNA mutations, the ETC proteins themselves are often structurally flawed due to the mutation. While SS-31 can stabilize the membrane environment, it cannot repair a fundamentally broken protein component. In contrast, the myopathy in patients with nDNA replisome defects often arises from secondary mtDNA instability (e.g., multiple deletions), but the ETC protein subunits encoded by the remaining mtDNA may be structurally normal. In this context, SS-31’s ability to organize these structurally sound proteins into more efficient supercomplexes could be highly effective. Therefore, the “failure” of MMPOWER-3 was not necessarily a failure of the drug, but rather a failure of a “one-size-fits-all” trial design that did not account for this critical genetic heterogeneity.

The NuPOWER Trial

This crucial learning has directly informed the path forward. Stealth BioTherapeutics has since initiated the NuPOWER trial, a Phase 3 study specifically designed to evaluate SS-31 in the nDNA-PMM patient population that demonstrated a positive response in MMPOWER-3.23 This trial, which is now fully enrolled, embodies the principles of precision medicine and represents a critical test of the refined therapeutic hypothesis generated from the previous study.29

Pathophysiology

Dry age-related macular degeneration (AMD) is a progressive retinal disease and a leading cause of irreversible blindness in older adults.14 Its pathogenesis is strongly linked to chronic oxidative stress and mitochondrial dysfunction within the high-energy-demand photoreceptors and the supportive retinal pigment epithelium (RPE).15 This bioenergetic failure leads to the progressive damage and death of these critical retinal cells, culminating in the loss of central vision, which is essential for tasks like reading and recognizing faces.14

The ReCLAIM-2 Trial

SS-31’s potential in this area was evaluated in the Phase 2 ReCLAIM-2 clinical trial, which randomized 176 patients with dry AMD and geographic atrophy (GA), an advanced form of the disease, to receive either SS-31 or placebo for 12 months.30 The trial did not meet its co-primary efficacy endpoints, which were based on functional measures of low-luminance best-corrected visual acuity (LL-BCVA) and the rate of GA lesion growth as measured by fundus autofluorescence.30

The Landmark Anatomical Finding

Despite the failure to meet the functional primary endpoints, the trial yielded a highly significant and promising result on a key anatomical endpoint measured by advanced retinal imaging (spectral-domain optical coherence tomography, or SD-OCT). Treatment with SS-31 resulted in a 43% reduction in the rate of photoreceptor loss compared to placebo, as quantified by the progression of Ellipsoid Zone (EZ) attenuation (nominal ).30 The EZ is a specific hyperreflective band visible on OCT imaging that corresponds to the mitochondria-rich layer of the photoreceptor inner segments.48 The integrity of the EZ is a direct structural surrogate for photoreceptor health, and its attenuation or loss precedes and predicts the loss of visual function.48

The ReCLAIM-2 results suggest that preserving the cellular machinery of vision (the photoreceptors) may be a more sensitive and reliable early measure of a neuroprotective drug’s effect than subjective and often variable functional tests. Vision loss in AMD is the direct result of photoreceptor death. The logical conclusion is that preserving the structure of these cells today is reasonably likely to predict the preservation of their function tomorrow.

The ReNEW and ReGAIN Program

Following these results, and in a landmark decision for the field, the FDA confirmed that EZ attenuation is an approvable primary clinical trial endpoint in dry AMD.52 This has paved the way for a large, global Phase 3 program, consisting of two identical pivotal trials named ReNEW and ReGAIN.15 These trials, which are currently enrolling patients, will use the rate of change in the macular area of photoreceptor loss (EZ attenuation) as their primary endpoint.23 The success of this program could validate a new paradigm for developing neuroprotective therapies in ophthalmology, where preserving retinal structure is accepted as a primary measure of therapeutic benefit.

IV. Comprehensive Safety and Tolerability Profile

Across a comprehensive clinical development program that has included multiple trials, diverse patient populations, and both short- and long-term administration, SS-31 has consistently demonstrated a favorable safety and tolerability profile.

4.1 Characterization of Adverse Events

The vast majority of reported adverse events (AEs) across all studies have been classified as mild to moderate in severity and have been transient in nature.25 The most frequently reported AEs are directly associated with the subcutaneous route of administration. These are injection site reactions, which can include localized erythema (redness), pruritus (itching), pain, swelling, bruising, and induration (hardening of the tissue).25 While common, these reactions are typically mild and manageable for most patients.

Systemic side effects have been reported less frequently and have also been generally mild. These can include headaches, dizziness, and abdominal pain.20 Importantly, extensive monitoring in clinical trials has not identified any clinically significant, treatment-related trends in vital signs, laboratory safety values (including hematology and clinical chemistry), or electrocardiogram (ECG) parameters.26 This favorable safety profile has been maintained even with long-term administration, as demonstrated in the TAZPOWER open-label extension, where patients received treatment for up to 168 weeks.26 The incidence of serious adverse events (SAEs) has been low across all studies, and those that have occurred have generally been deemed by investigators to be unrelated to the study drug.

4.2 Overall Risk-Benefit Assessment

The cumulative safety data from the SS-31 clinical program support a positive risk-benefit assessment, particularly for the patient populations being studied. The primary risks are associated with manageable, localized injection site reactions. For patients with severe, progressive, and life-limiting diseases such as Barth syndrome, primary mitochondrial myopathy, and dry AMD—for which there are few or no approved treatments—the potential benefits of a therapy that targets the core pathophysiology of the disease are substantial. The overall safety data suggest that these potential benefits may significantly outweigh the identified risks.

V. Synthesis and Future Directions: SS-31 as a Vanguard for Mitochondrial Medicine

SS-31 represents a paradigm shift in the approach to treating diseases of mitochondrial dysfunction. Its development journey, marked by both challenges and breakthroughs, has not only advanced a promising therapeutic candidate but has also generated invaluable lessons that will shape the future of mitochondrial medicine. Its story is a powerful illustration of how targeting a fundamental biological process can have broad therapeutic implications and how persistence in the face of complex clinical and regulatory hurdles can pave the way for innovation.

5.1 Key Learnings from the SS-31 Development Program

The legacy of SS-31 may ultimately be defined as much by the precedents it has set for how to develop drugs for complex diseases as by the therapeutic itself. The program has yielded several field-defining insights.

First, patient stratification is paramount. The MMPOWER-3 trial serves as a powerful cautionary tale against the use of heterogeneous “basket” trial designs in genetically diverse diseases. The trial’s overall negative result masked a clear signal of efficacy in a genetically defined subgroup. This outcome provides a definitive lesson: future success in mitochondrial medicine, and likely in many other complex genetic disorders, will depend on moving away from broad, phenotype-based populations toward meticulously stratified, genotype-specific therapeutic approaches.

Second, endpoint innovation is crucial for progress. The development programs in both dry AMD and Barth syndrome demonstrate that for slowly progressive and ultra-rare diseases, traditional functional endpoints may be insensitive or impractical. The regulatory acceptance of innovative anatomical (EZ attenuation in AMD) and intermediate (knee extensor strength in BTHS) endpoints can provide viable and more efficient paths to approval. This willingness to embrace novel, mechanistically sound surrogate endpoints is a major advancement that could accelerate the development of neuroprotective and restorative therapies across many fields.

Third, regulatory flexibility and patient advocacy are transformative forces. The protracted but ultimately successful regulatory saga for Barth syndrome is a testament to how a collaborative and persistent approach involving sponsors, patient communities, and regulators can overcome immense challenges in ultra-rare disease development. The journey from a “refusal to file” letter to an accelerated approval highlights the impact of compelling data from non-traditional sources like open-label extensions and the power of a unified patient voice in educating regulators about disease burden and unmet need.

5.2 The Future of SS-31

The future of SS-31 now hinges on the outcomes of its ongoing late-stage clinical trials. The NuPOWER trial, which is fully enrolled and focuses exclusively on the responsive nDNA-PMM patient subgroup, represents a critical test of the precision medicine hypothesis generated from MMPOWER-3. A positive result would not only provide a much-needed therapy for this specific patient population but would also be a resounding validation of the genotype-driven approach.

Similarly, the future in ophthalmology rests on the large, pivotal ReNEW and ReGAIN trials. Their success will depend on whether the promising anatomical improvements in photoreceptor preservation observed in Phase 2 can be replicated on a larger scale and confirmed as a valid surrogate for the long-term preservation of vision. For Barth syndrome, having secured accelerated approval, the challenge will be to fulfill the post-marketing requirements to verify clinical benefit in a confirmatory trial and to work toward potentially expanding the label to include younger and more severely affected patients who were not part of the pivotal study.

5.3 Broader Implications: A New Therapeutic Paradigm

The journey of SS-31, culminating in the first-ever FDA approval of a mitochondria-targeted therapeutic for Barth syndrome, serves as a landmark validation of an entire therapeutic strategy. It provides definitive proof-of-concept that targeting the fundamental bioenergetic machinery of the cell—specifically the cardiolipin-ETC supercomplex axis—is a viable and powerful approach to treating human disease.

This success has the potential to catalyze a new wave of investment and innovation in the development of therapeutics aimed at restoring mitochondrial health. By demonstrating that it is possible to pharmacologically stabilize the engine of the cell, SS-31 has opened the door to a new class of medicines. Such therapies hold immense promise not only for rare genetic mitochondrial diseases but also for a vast array of common, age-related conditions where mitochondrial decline is a key pathogenic driver, including heart failure, neurodegeneration, and metabolic syndrome. SS-31 may thus be remembered not only as a treatment for specific diseases but as the vanguard of a new and promising era of mitochondrial medicine.

Works cited

  1. www.khanacademy.org, accessed October 7, 2025, https://www.khanacademy.org/test-prep/mcat/biomolecules/krebs-citric-acid-cycle-and-oxidative-phosphorylation/a/oxidative-phosphorylation-the-major-energy-provider-of-the-cell#:~:text=Oxidative%20phosphorylation%20is%20the%20process,ATP%20synthase)%20to%20create%20ATP.
  2. Oxidative Phosphorylation – GeneGlobe – QIAGEN, accessed October 7, 2025, https://geneglobe.qiagen.com/us/knowledge/pathways/oxidative-phosphorylation
  3. Biochemistry, Oxidative Phosphorylation – StatPearls – NCBI Bookshelf, accessed October 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK553192/
  4. The Mechanism of Oxidative Phosphorylation – The Cell – NCBI Bookshelf, accessed October 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK9885/
  5. Programmed Cell Death (Apoptosis) – Molecular Biology of the Cell – NCBI Bookshelf, accessed October 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK26873/
  6. What is apoptosis, and why is it important? – PMC, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC1120576/
  7. Apoptosis – The Science Behind Cell Death – GeneGlobe – QIAGEN, accessed October 7, 2025, https://geneglobe.qiagen.com/us/knowledge/pathways/cellular-activity-metabolism-and-homeostasis-pathways/apoptosis
  8. Role of Cardiolipin in Mitochondrial Function and Dynamics in Health and Disease: Molecular and Pharmacological Aspects – PMC – PubMed Central, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6678812/
  9. The Role of Cardiolipin in Cardiovascular Health – PMC – PubMed Central, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4537736/
  10. Primary Mitochondrial Myopathies – Symptoms, Causes, Treatment | NORD, accessed October 7, 2025, https://rarediseases.org/rare-diseases/primary-mitochondrial-myopathies/
  11. Barth syndrome – Genetics – MedlinePlus, accessed October 7, 2025, https://medlineplus.gov/genetics/condition/barth-syndrome/
  12. tazpower – Barth Syndrome Foundation, accessed October 7, 2025, https://www.barthsyndrome.org/research/clinicaltrials/tazpower.html
  13. Mitochondrial Myopathy – Physiopedia, accessed October 7, 2025, https://www.physio-pedia.com/Mitochondrial_Myopathy
  14. Dry Age-related Macular Degeneration (AMD) – Prevent Blindness, accessed October 7, 2025, https://preventblindness.org/dry-age-related-macular-degeneration-amd/
  15. Stealth BioTherapeutics Announces Achievement of 50% Enrollment Target in Phase 3 ReNEW Study of Elamipretide in Patients with Dry Age-Related Macular Degeneration – PR Newswire, accessed October 7, 2025, https://www.prnewswire.com/news-releases/stealth-biotherapeutics-announces-achievement-of-50-enrollment-target-in-phase-3-renew-study-of-elamipretide-in-patients-with-dry-age-related-macular-degeneration-302401062.html
  16. Supercomplex – Wikipedia, accessed October 7, 2025, https://en.wikipedia.org/wiki/Supercomplex
  17. Structure of a mitochondrial supercomplex formed by respiratory-chain complexes I and III | PNAS, accessed October 7, 2025, https://www.pnas.org/doi/10.1073/pnas.0408870102
  18. Mitochondrial Respiratory Chain Supercomplexes: From Structure to Function – MDPI, accessed October 7, 2025, https://www.mdpi.com/1422-0067/23/22/13880
  19. Elamipretide (Brand Name FORZINITY™ for Barth Syndrome), accessed October 7, 2025, https://www.curefa.org/drug-development/elamipretide/
  20. Elamipretide Clinical Studies and Trials – Stealth BioTherapeutics, accessed October 7, 2025, https://stealthbt.com/programs-pipeline-copy/
  21. Natural history comparison study to assess the efficacy of elamipretide in patients with Barth syndrome – PMC – PubMed Central, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9438322/
  22. TAZPOWER Analysis: Elamipretide Significantly Improves Disease Symptomatology versus Natural History Controls in Barth Syndrome – Stealth BioTherapeutics, accessed October 7, 2025, https://stealthbt.com/wp-content/uploads/ASHG-2020_-TAZPOWER-72-wk-Poster_v2_10.1.2020_jas.pdf
  23. Elamipretide Clinical Trials – Stealth BioTherapeutics, accessed October 7, 2025, https://stealthbt.com/programs-pipeline/
  24. The mitochondrially targeted peptide elamipretide (SS-31) improves ADP sensitivity in aged mitochondria by increasing uptake through the adenine nucleotide translocator (ANT) – PMC, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10643647/
  25. TAZPOWER: Results of a Randomized, Double-Blind, Placebo-Controlled, Crossover and Open-Label Extension Trial of Elamipretide in Barth Syndrome – Stealth BioTherapeutics, accessed October 7, 2025, https://stealthbt.com/wp-content/uploads/MDA_2019_TAZPOWER_Results_Open-Label_Extension_of_Elamipretide_in_Barth_Syndrome.pdf
  26. 168-week open-label extension results of TAZPOWER : Research Summaries : Family Resources – Barth Syndrome Foundation, accessed October 7, 2025, https://www.barthsyndrome.org/barthsyndrome/familyresources/research-summaries/168-week-open-label-extension-results-of-tazpower.html
  27. Stealth BioTherapeutics Announces FDA Accelerated Approval of FORZINITY™ (elamipretide) injection, the First Therapy for Progressive and Life-limiting Ultra-rare Genetic Disease Barth Syndrome, accessed October 7, 2025, https://stealthbt.com/stealth-biotherapeutics-announces-fda-accelerated-approval-of-forzinity-elamipretide-hcl-the-first-therapy-for-progressive-and-life-limiting-ultra-rare-genetic-disease-barth-syndrome/
  28. Efficacy and Safety of Elamipretide in Individuals With Primary Mitochondrial Myopathy: The MMPOWER-3 Randomized Clinical Trial, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10382259/
  29. Mitochondrial Disease Therapeutic Pipeline – UMDF, accessed October 7, 2025, https://umdf.org/clinical-trials/therapeutic-pipeline/
  30. ReCLAIM-2: A Randomized Phase II Clinical Trial Evaluating Elamipretide in Age-related Macular Degeneration, Geographic Atrophy Growth, Visual Function, and Ellipsoid Zone Preservation – PubMed Central, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11599447/
  31. Stealth BioTherapeutics enrolls first patient in global Phase 3 clinical program for elamipretide in patients diagnosed with dry AMD | Ophthalmology Times, accessed October 7, 2025, https://www.ophthalmologytimes.com/view/stealth-biotherapeutics-enrolls-first-patient-in-global-phase-3-clinical-program-for-elamipretide-in-patients-diagnosed-with-dry-amd
  32. Barth Syndrome – BrainFacts, accessed October 7, 2025, https://www.brainfacts.org/diseases-and-disorders/neurological-disorders-az/diseases-a-to-z-from-ninds/barth-syndrome
  33. Barth syndrome – Orphanet, accessed October 7, 2025, https://www.orpha.net/en/disease/detail/111
  34. Barth Syndrome | Boston Children’s Hospital, accessed October 7, 2025, https://www.childrenshospital.org/conditions/barth-syndrome
  35. TAZPOWER: Biomarker Cardiolipin Ratios and Clinical Symptom Severity from a Randomized, Double-Blind, Placebo-Controlled – Stealth BioTherapeutics, accessed October 7, 2025, https://stealthbt.com/wp-content/uploads/UMDF_2019_TAZPOWER_Cardiolipin_Ratio_Results.pdf
  36. FDA Grants Accelerated Approval to Elamipretide, First Treatment for Barth Syndrome, accessed October 7, 2025, https://www.pharmacytimes.com/view/fda-grants-accelerated-approval-to-elamipretide-first-treatment-for-barth-syndrome
  37. After lengthy review, FDA snubs Stealth’s Barth syndrome therapy – FirstWord Pharma, accessed October 7, 2025, https://firstwordpharma.com/story/5967205
  38. elamipretide – Barth Syndrome Foundation, accessed October 7, 2025, https://www.barthsyndrome.org/advocacy/elamipretide.html
  39. FDA Advisory Committee Review of Elamipretide – Barth Syndrome Foundation, accessed October 7, 2025, https://www.barthsyndrome.org/advocacy/fdaadcomm.html
  40. Stealth BioTherapeutics Announces “Path Forward” Despite Disappointing Delay for Ultra-rare Barth Syndrome, accessed October 7, 2025, https://stealthbt.com/stealth-biotherapeutics-announces-path-forward-despite-disappointing-delay-for-ultra-rare-barth-syndrome/
  41. FDA Grants Accelerated Approval to First Treatment for Barth Syndrome, accessed October 7, 2025, https://www.fda.gov/news-events/press-announcements/fda-grants-accelerated-approval-first-treatment-barth-syndrome
  42. News & Events – Stealth BioTherapeutics Inc., accessed October 7, 2025, https://stealthbt.com/latest-news/
  43. Mitochondrial Myopathy – Children’s Hospital of Philadelphia, accessed October 7, 2025, https://www.chop.edu/conditions-diseases/mitochondrial-myopathy
  44. Primary Mitochondrial Myopathies – UMDF, accessed October 7, 2025, https://umdf.org/pmm/
  45. NuPower Study: A Phase 3 Randomized, Double-Blind, Parallel-Group, Placebo-Controlled Trial to Evaluate the Efficacy and Safety of Daily Subcutaneous Injections of Elamipretide in Subjects With Primary Mitochondrial Disease Resulting From Pathogenic Nuclear DNA Mutations (nPMD). – Orphanet, accessed October 7, 2025, https://www.orpha.net/en/research-trials/clinical-trial-network/626795?orphaCode=626795
  46. Dry Macular Degeneration – Symptoms and Treatments – AMDF, accessed October 7, 2025, https://www.macular.org/about-macular-degeneration/dry-macular-degeneration
  47. Understanding Dry Age-Related Macular Degeneration: A Patient’s Guide, accessed October 7, 2025, https://retinaassociatesofwny.com/understanding-dry-age-related-macular-degeneration-a-patients-guide/
  48. Challenges Associated With Ellipsoid Zone Intensity Measurements Using Optical Coherence Tomography – PMC – PubMed Central, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8543396/
  49. Ellipsoid Zone Defects in Retinal Vein Occlusion Correlates With Visual Acuity Prognosis: SCORE2 Report 14 | TVST, accessed October 7, 2025, https://tvst.arvojournals.org/article.aspx?articleid=2772433
  50. ELLIPSOID ZONE MAPPING AND OUTER RETINAL ASSESSMENT IN STARGARDT DISEASE, accessed October 7, 2025, https://www.oculogenetica.com/?mdocs-file=1122
  51. Longitudinal Ellipsoid Zone and Outer Retinal Integrity Dynamics Following Epiretinal Membrane Surgery – PubMed Central, accessed October 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8776576/
  52. Stealth BioTherapeutics Announces Achievement of 50% Enrollment Target in Phase 3 ReNEW Study of Elamipretide in Patients with Dry Age-Related Macular Degeneration, accessed October 7, 2025, https://stealthbt.com/stealth-biotherapeutics-announces-achievement-of-50-enrollment-target-in-phase-3-renew-study-of-elamipretide-in-patients-with-dry-age-related-macular-degeneration/
Scroll to Top