Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jan 6.
Published in final edited form as: Alzheimers Dement. 2020 Jan 6;16(1):131–143. doi: 10.1016/j.jalz.2019.06.4956

New Directions in Clinical Trials for Frontotemporal Lobar Degeneration: Methods and Outcome Measures

Adam L Boxer 1,*, Michael Gold 2, Howard Feldman 3, Bradley F Boeve 4, Susan L-J Dickinson 5, Howard Fillit 6, Carole Ho 7, Robert Paul 8, Rodney Pearlman 9, Margaret Sutherland 10, Ajay Verma 11, Stephen P Arneric 12, Brian M Alexander 13, Bradford C Dickerson 14, Earl Ray Dorsey 15, Murray Grossman 16, Edward D Huey 17, Michael C Irizarry 18, William J Marks 19, Mario Masellis 20, Frances McFarland 21, Debra Niehoff 5, Chiadi U Onyike 22, Sabrina Paganoni 23, Michael A Panzara 24, Kenneth Rockwood 25, Jonathan D Rohrer 26, Howard Rosen 1, Robert N Schuck 27, Holly D Soares 28, Nadine Tatton 5
PMCID: PMC6949386  NIHMSID: NIHMS1542150  PMID: 31668596

Abstract

INTRODUCTION:

Frontotemporal Lobar Degeneration (FTLD) is the most common form of dementia for those under 60 years of age. Increasing numbers of therapeutics targeting FTLD syndromes are being developed.

METHODS:

In March 2018 the Association for Frontotemporal Degeneration convened the Frontotemporal Degeneration Study Group (FTSG) meeting in Washington, DC to discuss advances in the clinical science of FTLD.

RESULTS:

Challenges exist for conducting clinical trials in FTLD. Challenges to be addressed are: 1) the heterogeneity of FTLD syndromes leading to difficulties in efficiently measuring treatment effects; and 2) the rarity of FTLD disorders leading to recruitment challenges.

DISCUSSION:

New personalized endpoints that are clinically meaningful to individuals and their families should be developed. Personalized approaches to analyzing MRI data, development of new fluid biomarkers and wearable technologies will help to improve the power to detect treatment effects in FTLD clinical trials and enable new, clinical trial designs as leveraged from the experience of oncology trials. A computational visualization and analysis platform that can support novel analyses of combined clinical, genetic, imaging, biomarker data with other novel modalities will be critical to the success of these endeavors.

Introduction

Frontotemporal Lobar Degeneration (FTLD) is the neuropathological term for a related group of rare neurodegenerative disorders that cause a spectrum of impairments in personality, cognitive ability, language, and motor function. These include behavioral variant frontotemporal dementia (bvFTD), primary progressive aphasias (PPA) and the parkinsonian disorders, corticobasal syndrome (CBS) and progressive supranuclear palsy (PSP). At present there are no approved symptomatic or disease modifying treatments for FTLD. Medications that are approved for use in other diseases are often used to manage FTLD symptoms without lasting success, but none have been found to slow or stop the progression of FTD [1-3]. Current management for FTLD relies on these symptomatic therapies as well as non-pharmacological interventions that include: reduction of excess stimulation from the environment combined with management of inappropriate or repetitive behaviors using tailored activities programs [4, 5], language retraining or speech therapy where possible [6, 7], and the use of physiotherapy and occupational therapy aids and modifications to the home environment to support progressive loss of motor skills [8]. These interventions offer partial but temporary symptomatic relief, address some of the caregiver burden but do not substantially alter the course of this fatal spectrum of disease. Later disease stages often require institutional care where behavioral problems, mutism, parkinsonism and dysphagia are managed symptomatically.

The Frontotemporal Degeneration Treatment Study Group (FTSG), a program of the Association for Frontotemporal Degeneration (AFTD), was founded in 2010 to promote collaborations between academic and pharmaceutical industry researchers focused on drug development for FTLD and related disorders [2, 9]. Since the last FTSG meeting that took place in 2016, much progress has been made in therapeutically relevant FTLD research. With increasing numbers of potential therapies entering familial FTLD (f-FTLD) clinical trials, the FTSG organized a meeting in Washington, DC, March 2018 in partnership with the National Institute of Neurological Disorders and Stroke, to discuss clinical trial methodology and outcome measures for the FTLD spectrum of disorders. Two key challenges to FTLD clinical trial design were identified as topics for this meeting: 1) the heterogeneity of clinical symptoms in FTLD syndromes caused by the same mutation or underlying pathology, leading to difficulties in efficiently measuring treatment effects using clinical or imaging outcome measures; and 2) the rarity of FTLD disorders leading to recruitment challenges and the necessity for trial designs and instruments that can optimize the measurement of treatment effects in small trial samples. This manuscript summarizes the presentations and discussion from that meeting and highlights new strategies to improve FTLD drug development.

Clinical trial design in rare FTLD disorders

The complexity of FTLD phenotypes and range of syndromes creates a significant challenge for clinical trial design, along with the fact that the FTLD disorders are considered rare diseases (less than 200,000 affected in the US). Collecting true population-based estimates for FTLD disorders is problematic given the limited public awareness of this younger onset dementia, clinical presentations that can overlap with other diseases, and the absence of validated biomarkers to distinguish FTLD from other neurological and psychiatric disorders. A recent study in the UK [10], reported a combined prevalence of 10.8 per 100,000 for bvFTD, PPA, PSP and CBS for all ages (40-100 years) with a peak between 65-70 years of approximately 45 per 100,000 which is consistent with previous prevalence estimates for FTD and PPA [11, 12]. Interest in participation in clinical trials is very high among familial FTLD kindreds as well as families living with sporadic FTLD, which has facilitated a number of multi-site clinical trials for FTLD disorders including bvFTD, semantic variant PPA (svPPA) and multiple studies in PSP [13-16]. Greater than 85% of participants in a survey for the Advancing Research and Therapies in Frontotemporal Lobar Degeneration (ARTFL) project, described below, indicated a strong interest to participate in a clinical trial.

There have been few randomized, placebo controlled trials in FTLD (3). Previous clinical trials have demonstrated the feasibility of using behavioral questionnaires, cognitive scales and functional activity ratings as outcome measures. Although no study to date has yielded evidence of therapeutic efficacy, previous trials have laid the groundwork for sharing data that could improve trial design [17]. Previous trials may have been unable to detect treatment effects for a number of reasons such as outcome measures that do not address clinical, etiological and imaging heterogeneity between patients carrying the same molecular diagnosis, inadequate sample size, and participants being too late in the course of the disease to demonstrate benefit. Refining FTLD patient selection and trial design will gain even greater importance as new disease-modifying therapeutics are developed [17]. The two largest industry sponsored trials in bvFTD () and FTLD due to progranulin gene mutations (FTLD-GRN; ) have not yet been published, and it is anticipated that data shared from these studies would advance our understanding of trial design for FTLD. Stronger mechanisms to ensure prompt publication and data sharing, based on the Collaboration for Alzheimer’s Prevention (CAP) principles [18], will be particularly important for a rare disease and need to be incorporated into future FTLD clinical trials.

Despite these challenges, new treatments targeting tau gain of function, progranulin haploinsufficiency and Chromosome 9 open reading frame 72 (C9orf72) hexanucleotide repeat expansions are progressing in clinical development for FTLD and related disorders, with some agents such as anti-tau monoclonal antibodies having entered large-scale efficacy studies for PSP ( and ). Table 1 summarizes drugs recently tested, in late stages of preclinical development, or currently under active evaluation. These ongoing and planned clinical trials across the spectrum of FTLD highlight the urgency of developing novel outcome measures, patient stratification tools and clinical trial designs as proposed in this project. Therapies that leverage or modify the immune system to treat FTLD are now in clinical trials. Tau immunotherapies are being tested by several groups who are leveraging the clinical homogeneity of patients with PSP-Richardson’s syndrome [16, 19] or non-fluent variant PPA [20], which are considered “pure” 4 repeat tauopathies with well-defined natural history of disease progression. These FTLD syndromes provide cohorts in whom it may be easier to demonstrate, and hopefully define, clinically meaningful endpoints that could achieve regulatory approval. A trial of a monoclonal antibody that blocks a progranulin receptor, and thereby hypothesized to increase progranulin levels, is also now underway (Table 1).

Table 1.

Potential FTLD Therapeutics

Drug Mode of Action Status Ref NCT*
GRN-targeted therapeutics
FRM-0334 HDAC inhibitor Phase 2 (negative) n/a 01835665
Chloroquine Vesicular pH modulator Repurposed [108] -
Nimodipine Increased progranulin secretion Repurposed; phase 1b (neg) [109] 01835665
AL-001 Anti-sortilin mAb Phase 1 n/a 03636204
Proprietary A, B HDAC inhibitor Preclinical [110] -
Proprietary A-C AAV gene therapy Preclinical [111] [112] -
C9orf72 therapeutics:
Proprietary A, B C9orf72 antisense oligos Phase 1 ALS; FTLD planned [113, 114] 03626012
Tau-targeted therapeutics:
LMTX (Methylene Blue) Protein clearance activator Phase 3 (neg. for bvFTD) n/a 01626378
Lithium carbonate GSK inhibitor Phase 2 FTD n/a 02862210
Abeotaxane (TPI-287) microtubule stabilizer Phase I (neg. for CBD, PSP) n/a 01966666
Salsalate Tau acetylation inhibitor Phase 1 PSP; abandoned [115] 02422485
ABV-8E12 N-terminal anti-tau mAb Phase 2 PSP [116] 02985879
BIIB092 N-terminal anti-tau mAb Phase 2 PSP [117] 02460094
BIIB092 N-terminal anti-tau mAb Phase 1b: CBD, nfvPPA, sMAPT [117] 03658135
AADvac1 Active anti-tau vaccine Phase 1: nfvPPA [118] 03174886
UCB0107 Mid-domain anti-tau mAb Phase 1 [119] -
ASN001 o-GlcNACase inhibitor Phase 1 [120] -
IONIS-MAPTrx Antisense oligonucleotide Phase 1 AD [121] 03186989
Other (Immunomodulatory, neuroprotective therapeutics):
NP001 Macrophage activation inhibitor Phase 2 ALS negative [122, 123] 03186989
DLZ Kinase inhibitor Neuroprotective agent Phase 1 ALS [124] 02655614
Palliative Approaches:
Oxytocin Symptomatic improvement Phase 2 bvFTD [125] 01386333
Rivastigmine Cholinesterase inhibitor Phase 2 PSP n/a 02839642
Transcranial DC stim Electric current stimulation N/A (pilot) bvFTD, PPA [126] 02999282
Transcranial magn. stim Magnetic field stimulation PPA [127] 03406429

Antisense oligonucleotide (ASO) therapy has been demonstrated to be effective in the central nervous system when used to treat spinal muscular atrophy [21, 22]. Oligonucleotides offer the opportunity for precision design with a sequence and modifications that can improve their selectivity, stability and specificity. Current platforms create either a stereo-random mixture of oligonucleotides, or more recently a pure stereo-isomer [22]. Two different ASO programs targeting the C9orf72 mutation are approaching the clinical stage for FTLD and an anti-MAPT ASO trial is underway in AD. This ASO could also potentially be used to treat FTLD due to MAPT mutations in the future.

Studies of FTLD syndromes using clinical endpoints and volumetric MRI provide a measure of disease progression and indicate that many FTLD syndromes (bvFTD, CBS, PSP) progress more rapidly than AD thereby enabling smaller and shorter trials and the potential to learn from successes and failures more quickly [23]. Clinical trials that enroll presymptomatic familial FTLD mutation carriers have the potential to act as ‘prevention’ studies, but are also more dependent on the development of highly predictive biomarker or clinical outcomes in a reasonable period of time following the model of the Dominantly Inherited Alzheimer’s Network Treatment Unit (DIAN-TU) trials [24, 25]. New FTLD natural history studies are beginning to develop similar capabilities.

The role of natural history studies in FTLD

In 2013, the National Alzheimer’s Project Act–Alzheimer’s Disease Related Dementias Summit identified key research priorities for FTLD [26]. With an ultimate goal of developing effective therapies for FTLD, the clinical research priorities included the formation of a clinical trials ready research network and development of new biomarkers for FTLD. The ARTFL network, created in 2014, is a large cross sectional and natural history study of sporadic FTD disorders in the US and Canada. Fully integrated with this program is the LEFFTDS (Longitudinal Evaluation of Frontotemporal Dementia Subjects) project, a longitudinal observational study of autosomal dominant FTLD-causing mutation families (C9orf72, GRN or MAPT), with a focus on developing pre-symptomatic biomarkers for FTLD (Boeve et al., personal communication). The GENFI (Genetic Frontotemporal Dementia Initiative) consortium includes sites in Europe and Canada that follow FTLD mutation carriers with the objective of finding diagnostic and disease progression markers. More robust natural history data from all FTLD syndromes is needed in order to develop clinically meaningful outcome measures and to better inform drug development for both symptomatic and disease modifying therapies. Functional and quality of life outcomes may provide opportunities to capture clinically meaningful outcome measures for a broad variety of FTLD phenotypes but there are few such outcome measures at this time that are FTLD specific. A better understanding of how persons diagnosed with FTLD and their caregivers would define meaningful functional stabilization or improvements that impact quality of life is needed [27, 28]. Additionally, what constitutes a clinically meaningful benefit for asymptomatic or questionably symptomatic mutation carriers is not agreed upon.

Like the LEFFTDS network, the GENFI network also follows familial FTLD kindreds with a goal of developing multi-modal MRI and fluid biomarkers and genomics methods to identify predictive factors, neuroanatomic correlates and variability in the natural history of disease progression [19, 29, 30]. By focusing on asymptomatic or mildly symptomatic f-FTLD patients who have relatively little neuropathology, future clinical trials should have improved power to detect treatment effects of these new therapies.

Heterogeneity of FTLD syndromes and outcome measures: New approaches to measuring disease progression

FTLD encompasses an array of clinical syndromes involving behavior, speech and/or motor deficits that arise from a handful of similar underlying brain pathologies, most commonly FTLD-tau or FTLD-TDP [31, 32]. The clinical course of FTLD generally begins as one of the distinct phenotypic variants and often progresses to involve other cognitive, behavioral and motor domains [33]. Survival ranges from 2-13 years after diagnosis (depending on clinical syndrome and underlying pathology), but averages about 8-10 years [34]. Slower progression cases with longer survival (ranging 20-30 years) have been described [35, 36]. Existing clinical instruments like the Neuropsychiatric Inventory may help classify subtypes within a particular syndromic diagnosis such as behavioral variant FTD [37] but cannot identify the underlying molecular pathology causing the syndrome [38]. Volumetric MRI is currently the best available technology at an individual level for the in vivo identification of neuron loss in FTLD, although the neuropathological correlates of MRI defined brain atrophy have not been fully validated [39]. Resting state fMRI can identify abnormalities in presymptomatic mutation carriers [40] but FDG PET may be more promising for capturing disease progression [41]. Emerging data demonstrate the correlation of bvFTD subtypes with distinct patterns of degeneration [42, 43] and provide a potential network-based model of the various phenotypes [44]. Furthermore, data driven approaches applied to volumetric MRI from genetic FTLD also shows promise for identifying different FTLD syndromes [45, 46]. MRI-based imaging measures such as voxel-based morphometry, Diffusion Tensor Imaging (DTI) and arterial spin label perfusion change over time in individual FTLD patients and generally show good correlations with clinical measures [47]. A challenge is that the data acquired from these images are often highly variable across syndromes caused by the same underlying pathology, but also even within the same clinical FTLD syndrome. Ideally an imaging method would provide a way of following an individual patient’s atrophy patterns regardless of FTLD syndrome to predict or distinguish their variable trajectory.

MRI-based approaches to account for heterogeneity within FTLD syndromes.

The underlying phenotypic heterogeneity of FTLD clinical syndromes argues for a personalized medicine approach able to capture individualized measures of change based on the patient’s baseline phenotype. A new imaging approach being investigated is the use of W-score maps that highlight how each individual voxel’s W-score (similar to Z-score, corrected for demographic variables) in FTLD images differ from those in normal brains, allowing quantification of the total burden or pattern of atrophy and assigning scores based on these maps which clearly differentiate FTLD-CDR=0 from FTLD-CDR=1 or higher [46, 48]. These maps may aid in the visualization of early neurodegenerative change; however, more data sets from younger healthy controls will be required in order to understand the observed variations in the rate of change. Increasingly, MR imaging is being combined with putative fluid biomarkers in an effort to stage and monitor FTLD with prediction of progression through a multi-modal approach [49-51].

A new, multidomain, global rating scale to measure clinical heterogeneity.

The LEFFTDS and ARTFL networks have developed a new scale based on the FTLD-CDR [48] that incorporates motor and sensory domains as well as separate streams of information for patients, informants and neuropsychologists, called the Multidomain Impairment Rating (MIR) scale as a global and quantitative clinical burden rating scale (Boeve et al., personal communication). The MIR is designed to be more sensitive than standard scales to the earliest signs and symptoms of FTLD in mutation carriers. Using standard lobar volumetric assessments, volumetric MRI in MAPT and other f-FTLD kindreds demonstrate prominent atrophy rates in symptomatic carriers, intermediate rates in asymptomatic carriers and only age related changes in non-carriers [52]. Modeling such rates of decline across different imaging modalities in mutation carriers at different MIR-defined stages of disease, may help to understand phenoconversion from clinically asymptomatic to symptomatic FTLD. A better understanding of the onset, duration and variability of this window could also lead to the identification of biomarkers that can predict or measure this change. The MIR will likely be an important tool to timestamp phenoconversion, a necessary step in biomarker validation.

Fluid biomarkers.

There is a growing literature on cerebrospinal fluid (CSF) and blood neurofilament light chain (NfL), viewed as a biomarker of neurodegeneration [53-55] and as a candidate marker of disease onset in FTLD. Furthermore, it may serve as a prognostic biomarker for genetic and sporadic FTLD [56-58] and reflect disease severity and rate of progression in some sporadic FTLD subtypes [54, 59-61]. Recent biomarker development studies reflect a growing trend to create test panels with a combination of a large number of analytes to provide a foundation for discrimination between clinically defined syndromes within FTLD and other neurodegenerative diseases such as AD and Amyotrophic Lateral Sclerosis/Motor Neuron Disease (ALS/MND) disorders [56, 62-64]. However, a weakness of this approach is that many previous efforts employing statistically clustered combinations of fluid biomarkers have often failed to replicate. Other potential fluid biomarkers that reflect changes in autophagy, neuroinflammation, RNA metabolism and mitochondrial function are a growing area of study in FTLD and other dementias [64]; however, it is not well understood whether this broader spectrum of measures will reflect early neurodegenerative processes or late responses to neurodegeneration.

Relating these biomarkers to the accumulation of insoluble deposits of tau and/or TDP-43 measured at autopsy in FTLD will be important. Even the relationship of TDP-43 and tau deposition to the onset and progression of sporadic FTLD syndromes is not well understood. For example, other than in MAPT or TARDBP mutation carriers, it is not known whether changes in these proteins initiate, mediate, contribute to or simply reflect other processes in disease progression. The complexity of biomarker discovery and validation for various heterogeneous FTLD syndromes in comparison to the simpler and more pathologically and clinically homogeneous AD syndromes has resulted in fewer specific biomarkers, and no pre-symptomatic biomarkers of sporadic disease. This makes it more challenging to develop a biological definition for FTLD, as has been recently suggested for AD [65]. Similarly, applying the recent FDA draft guidance for prodromal AD drug development based on fluid or imaging biomarkers [66] represents a higher hurdle for prodromal FTLD. Nevertheless, with the strong data already obtained using CSF and blood NfL, use of this fluid biomarker to define or predict onset of clinical symptoms may enable FTLD prevention trials in asymptomatic or early symptomatic FTLD mutation carriers. In such a scenario, the time to elevation in blood NfL or the rate of increase of NfL concentration in the late pre-symptomatic stage of disease or even change from baseline could be used as potential endpoints for prevention trials (Table 2). Such a scenario will require that blood NfL levels do indeed reflect underlying neurodegeneration and are strongly predictive of future clinical status allowing them to be validated as a surrogate endpoints as has been done in other diseases such as HIV, in which some clinical trials have relied on a surrogate biomarkers that predicts future disease for approvals [67].

Table 2a:

Draft FDA Guidance for Approvals in Presymptomatic/Early AD

Stage 1 Stage 2 Stage 3 Stage 4
Preclinical Prodromal (MCI) Early AD Mild-moderate AD
Definition
  • Asymptomatic

  • Biomarker evidence of pathology (only)

  • Detectable cognitive changes

  • No functional impairment

  • Cognitive impairment

  • Mild functional impairment

  • Overt dementia

  • Cognitive and functional impairment

Possible endpoints
  • Biomarker

  • Imaging

Cognitive scale(s) only (biomarker supported dx) Clinical scale(s) to assess both daily function and cognitive effects Clinical scale(s) to assess both daily function and cognitive effects
Clinically meaningful effect for approval? Not required Clinically meaningful ideal; not required Clinically meaningful effect required Clinically meaningful effect required
Approval type Conditional approval Conditional approval Regular approval Regular approval

Autosomal dominant FTLD and sporadic FTLD – the same disease?

The autosomal dominant FTLD gene mutations afford a unique insight into the molecular ‘switches’ that convert asymptomatic to symptomatic mutation carriers. It is hoped that the biology of this prodromal transition will also provide new insight into the causes and earliest biological changes in sporadic FTLD. While the autosomal dominant gene mutations provide greater confidence for an FTLD diagnosis and can help to assure recruitment of the right patients into clinical trials, it is not clear how different FTLD causing mutations lead to biochemical changes that converge on the same brain networks that produce the unique phenotypes associated with FTLD. Further, while insights based on the study of f-FTLD are often relied on for drug discovery, it is not known how such genetic FTLD syndromes relate to sporadic FTLD or how findings developed in preclinical models based on a particular f-FTLD mutation (such as P301S MAPT) will relate to other genetic (such as V337M MAPT) FTLD patients. Initial data from bvFTD patients carrying mutations in C9orf72, GRN or MAPT suggest that they are very similar from a clinical and MR imaging perspective to sporadic FTLD patients (Heuer et al., submitted A&D).

An important question is when (and where) neurodegeneration in FTLD begins? In autosomal dominant FTLD, mutations are present from conception [68] and recent data in C9orf72 mutation carriers suggest there is a lifelong propensity to develop psychiatric disorders. Further, each gene demonstrates heterogeneity in its associated clinical syndromes, and family members with the same mutation may present with a different clinical syndrome [69] (M. Ramos et al., personal communication). MAPT mutations most often lead to a bvFTD phenotype, but may be expressed as the movement disorder syndromes of PSP or CBS. With more than 60 mutations and a small number of affected families, trying to map the different MAPT mutations to different brain networks is daunting [70, 71]. GRN and C9orf72 mutations offer similar challenges with C9orf72 providing additional variability with of a mix of clinical syndromes that may be bvFTD, or ALS, or FTD with ALS, or ALS with a range of behavioral or cognitive impairment or with CBS or nfvPPA [72-74]. To best understand these processes, combining data from genetic and sporadic FTLD patients may be necessary. For example, a recent publication examined the overlap between ALS and FTLD revealing a number of novel loci and functional pathways shared by ALS, bvFTD and PSP and that the MAPT H1 haplotype conferred risk for ALS [75]. Together, these studies suggest that studying both autosomal dominant and sporadic FTLD syndromes in parallel, with the same clinical, imaging and biomarker tools will help to overcome limitations of studying one population on its own, thereby increasing the likelihood of progress towards an effective therapy.

Developing targeted therapies for molecularly defined subsets of a disease

The FDA has recently issued draft guidance on “Developing Targeted Therapies in Low-Frequency Molecular Subsets of a Disease. Guidance for Industry” [76]. This guidance was issued to address challenges in the development of targeted therapies for diseases with multiple molecular subsets, when some of these subsets are too small to deliver robust and conclusive data. For these targeted therapies, moving forward with drug development towards approval is challenged by specific patient recruitment, interpretation of results and extrapolating findings to putatively similar molecular subtypes [76]. The new guidance recommends that grouping patients with different molecular alterations into a single trial may be based on a scientific rationale that the grouped patients will have a similar pharmacological response to a new drug. This would allow for the possibility of extrapolating efficacy findings across multiple subsets in spite of a low number of patients in some subsets. Although the guidance is focused on developing targeted therapies in low-frequency subsets within a single disease, some principles may be applicable to basket trial designs where more than one disease is included in a single clinical trial [77]. One such basket design clinical trial is now underway with an anti-tau monoclonal antibody in FTLD-tau syndromes () and other similar studies in FTLD-TDP syndromes are planned.

Precision medicine has advanced in oncology by classifying many cancers by the presence of known pathogenic gene mutations; allowing for inclusion of additional patients in trials based on the presence of a specific genetic marker in their cancerous cells [78, 79]. This ability to identify subpopulations that may respond to a specific treatment, and tailor treatment to the individual characteristics of each patient based on biomarkers, has contributed to an understanding of trial design elements that could also be applied to FTLD. In oncology, platform trials using master protocols with multiplexed biomarkers improve the efficiency of testing novel agents and allow for the use of common controls, thereby reducing overall sample sizes necessary to test multiple new drugs. Adaptive trials use the accumulating data to support decision-making on modifying a study in a pre-specified manner such as dropping arms, using surrogate endpoints or adaptive randomization and Bayesian analysis [80, 81]. For example, therapeutics for glioblastoma are limited but molecular knowledge of the disease is significant. The INdividualized Screening trial of Innovative Glioblastoma Therapy (INSIGhT) [82] and the Adaptive Global Innovative Learning Environment for Glioblastoma (GBM-AGILE) were devised as multi-arm platforms to support and inform drug development using biomarkers that allow for accumulating trial data to identify possible responders [83, 84]. As increasing numbers of outcome and pharmacodynamic biomarkers are developed for FTLD, similar approaches might be pursued.

Personalized endpoints, data sharing and new technologies

Personalized clinical outcomes, in which the clinical outcome may vary between different patients in an effort to measure the most important and relevant signs, symptoms, functions, as well as the degree of severity of these impairments in each individual, are one approach to capturing heterogeneous changes in diseases caused by a common underlying pathology [85, 86]. Such personalized outcomes are encouraged by the FDA’s Patient-Focused Drug Development initiative [87]. Approaches to the development of personalized outcomes include the ‘most bothersome symptoms’ (MBS) approach [85], Goal Attainment Scaling [88], and Computer Adaptive Testing (CAT) [89]. Goal attainment scaling (GAS) is an example of how a quantitative approach to measuring individual outcomes can be developed within a structured method for documenting patient-centered problems and care [90]. The benefits of GAS are the improvement in stakeholder engagement and empowerment of the patient, caregiver and clinician; as well as providing inherent clinical meaningfulness in capturing preferences [91]. It has been used successfully in AD clinical trials (ACADIE, VISTA) demonstrating GAS scores were more responsive than standard outcomes including the ADAS-Cog and the CIBIC+ [92-94]. Other studies have subsequently determined that GAS can help caregivers of dementia reach their own goals via GAS [95] and that other platforms such as the Hierarchy Model of Needs in Dementia have value in relating needs to individual goal setting instruments for patients and caregivers [96].

There is increased demand for broader data sharing by research funders and the recognition of a secure environment to store such data and make it available for analysis within the disease subset as well as externally to other diseases and potentially other data platforms. The limited capabilities of existing platforms that serve to disseminate pre-clinical and clinical data such as the National Alzheimer’s Coordinating Center-FTLD Module (NACC-FTLD), Laboratory of Neuroimaging (LONI) and Database of Genotype and Phenotype (DbGAP), suggest that more fit for purpose platforms for multimodal data sharing for FTLD will be needed. Other drivers include the evolution of wearable devices and the use of mobile technology to record, store and transmit user produced data, creating a “digital phenotype” that can be uploaded and analyzed as part of clinical data collection, most notably in the Parkinson’s disease field [97-99]. Database challenges include: ensuring data privacy and security, gaining regulatory approval of remote tracking devices, extracting the maximal amount of information from the smallest number of devices and locations and validating outputs against existing standards, as well as providing sites that can not only store data but provide a cloud-based platform for data analysis with large data sets. The NIH ‘Accelerating Medicines Partnership’ program for Parkinson’s disease is a public-private partnership that seeks to address this challenge by creating a cloud-based resource that can store and analyze complex datasets for fluid biomarkers in patient and control populations. A similar effort could be developed with NIH for FTLD, or a focused precompetitive alliance of partners from industry, patient advocacy organizations, and philanthropy could accelerate this effort as done for Alzheimer disease (https://c-path.org/programs/cpad/).

Essential to the success of remote data collection and the creation of a shared database is concise informed consent to increase data and biospecimen access [100]. Critical to the success of any database is well-curated data and well defined data standards [101, 102] that can tease apart symptoms and signs that may be common across different diseases or subtypes. Such databases can transform clinical trials with high frequency, objective and continuous data [103]. Developing a sustainable ecosystem that captures remotely tracked, continuous, biometric data will require a collaborative effort across many groups of stakeholders as demonstrated for AD with the Coalition to Prevent Alzheimer’s Disease (CPAD) and Global Alzheimer’s Association Interactive Network (GAAIN) databases, and Pooled Resource Open-Access ALS Clinical Trials Database (PRO-ACT) for ALS [104-106]. Well-curated databases can speed the pace and reduce the cost of drug development by creating data standards that can aid in the evaluation of efficacy and safety of new therapies. They have the potential to be reviewed and qualified by the FDA as a ‘drug development tool’; but to be successful requires buy-in across all stakeholders with relevant drug development pipelines.

Conclusions and future directions

Increasing numbers of clinical trials for FTLD are planned in the next few years. Particularly exciting are therapies targeting altered levels or mutant forms of products from the FTLD causing genes, C9orf72, GRN and MAPT. In addition, the successful enrollment of large clinical trials of anti-tau therapies in PSP are likely to enable new clinical trials of these therapies in sporadic FTLD syndromes with predicted underlying 4R tau pathology including nfvPPA and corticobasal syndrome.

Many challenges remain to finding efficacious therapies for FTLD. Further development of statistical and biomarker approaches to account for heterogeneity of phenotypes in both genetic and sporadic FTLD syndromes will be necessary. One potential solution is to develop personalized endpoints to measure treatment effects. These personalized endpoints may have increased clinical meaningfulness if approaches such a goal attainment scaling are used as a basis for endpoint development.

While a strong body of evidence now exists to support the use of blood or CSF NfL as a fluid biomarker to help define disease onset and severity of neurodegeneration, new biomarkers that can be deployed in asymptomatic FTLD mutation carriers or questionably symptomatic individuals with sporadic forms of FTLD will be necessary. With new FDA draft guidance for approval of drugs to prevent dementia in asymptomatic individuals who are at risk for disease, such biomarkers will be increasingly important in the future.

Novel clinical endpoints, possibly acquired through new wearable and other mobile technologies may further increase sensitivity and power to detect treatment effects, and might also be sensitive to early features of disease prior to the onset of overt clinical symptoms [107]. To make best use of these novel technologies, improved technological infrastructure and ironclad policies to ensure sharing of clinical and biomarker data and remaining biological specimens from completed clinical trials will also be necessary. Efforts to incorporate such policies into new treatment trials facilitated by or conducted within the North American ARTFL/LEFFTDS consortium and the European and Canadian GENFI project are an important first step to an improved publication and data sharing approach for FTLD clinical trials. While there is much work to be done, the rapid pace of clinical therapeutic development for FTLD bodes well for the imminent development of effective therapies.

Table 2b:

Application of Draft Early AD Approval Guidance to FTLD

Stage 1 Stage 2 Stage 3 Stage 4
Population Preclinical (mut. carriers) Prodromal (MCI/MBI) Early dementia Mild-moderate disease
FTLD-CDR FTLD-CDR = 0 FTLD-CDR = 0.5 FTLD-CDR = 1.0 FTLD-CDR > 1.0
Definition
  • Asymptomatic

  • Biomarker evidence of pathology (only)

  • Questionable or mild clinical disease

  • No functional impairment

  • Clinical impairments

  • Mild functional impairment

  • Overt dementia

  • Clinical or functional impairment

Possible endpoints Biomarker
  • NfL Imaging

  • atrophy

Clinical scale ± Biomarker Clinical scale(s) to assess both daily function and clinical effects Clinical scale(s) to assess both daily function and cognitive effects
Clinically meaningful effect for approval? Not required Clinically meaningful ideal; not required Clinically meaningful Clinically meaningful
Approval type Accelerated Accelerated Accelerated Accelerated
  1. Systematic review: The authors reviewed the literature using traditional (e.g., PubMed) sources, meeting abstracts and presentations. There have been a limited number of randomized placebo controlled clinical trials performed in frontotemporal lobar degeneration syndromes in the past. A variety of endpoints have been used in these studies; all were negative. The relevant citations are appropriately cited.

  2. Interpretation: A variety of challenges exist for conducting clinical trials in FTLD. Most prominently, these are: 1) the heterogeneity of FTLD syndromes leading to difficulties in efficiently measuring treatment effects using common clinical or imaging outcome measures; and 2) the rarity of FTLD disorders leading to recruitment challenges and difficulties with adequate power to detect treatment effects.

  3. Future directions: A limited number of clinical trials are underway and more are planned for both familial and sporadic FTLD syndromes. New personalized endpoints that are most clinically meaningful to individuals and their families should be developed. Additionally, more powerful approaches to analyzing clinical and MR imaging data, development of new fluid biomarkers and wearable technologies will help to improve the power to detect treatment effects in FTLD clinical trials and enable new, more efficient clinical trial designs modeled on oncology. More widespread sharing of clinical trial data and biofluid samples will be critical to developing new endpoints and refining FTLD clinical trial designs.

Acknowledgements

This work was funded by the Association for Frontotemporal Degeneration and the NIH (U54NS092089, R01AG038791, U01AG045390)

Competing Interests

Adam L. Boxer: receives research support from the NIH (U54NS092089, R01AG038791, U01AG045390), the Tau Research Consortium, the Association for Frontotemporal Degeneration, Bluefield Project to Cure Frontotemporal Dementia, Corticobasal Degeneration Solutions, the Alzheimer’s Drug Discovery Foundation and the Alzheimer's Association. He has served as a consultant for Aeton, Abbvie, Alector, Amgen, Arkuda, Eisai, Ionis, Ipierian, Janssen, Lundbeck, Merck, Novartis, Samumed, Toyama and UCB, and received research support from Avid, Biogen, BMS, C2N, Cortice, Eli Lilly, Forum, Genentech, Janssen, Novartis, Pfizer, Roche and TauRx.

Michael Gold: is a full-time employee of AbbVie.

Howard Feldman: receives research funding from the NIH; ADCS Study U19AG10483-26, Canadian Institutes of Health Research #287674 and #363926 with Weston Foundation, Biohaven Pharmaceuticals, Toyama Pharmaceuticals, and development grant funding from Probiodrug. He has UCSD service agreements with Axon Neurosciences, Eisai Pharmaceuticals, Genentech/Roche Pharmaceuticals, Banner Health Institute, Samus Therapeutics, Merck Pharmaceuticals, Tau RX, Arkuda Therapeutics, and Samumed; and has received travel expenses from Axon Neurosciences, Alion Pharmaceuticals, Probiodrug, and Dominantly Inherited Alzheimer's Disease. He serves on the Scientific Advisory Board of the Tau Consortium.

Bradley F. Boeve: has served as an investigator for clinical trials sponsored by GE Healthcare and Axovant. He receives royalties from the publication of a book entitled Behavioral Neurology Of Dementia (Cambridge Medicine, 2009, 2017). He serves on the Scientific Advisory Board of the Tau Consortium. He receives research support from NIH, the Mayo Clinic Dorothy and Harry T. Mangurian Jr. Lewy Body Dementia Program and the Little Family Foundation

Susan L-J Dickinson: on staff at the Association for Frontotemporal Degeneration and a member of the National Institute for Neurological Disorders and Stroke Advisory Council.

Howard Fillit: has been a consultant to: Axovant, vTv, Lundbeck, Otsuka, Lilly, Biogen, Roche, Genentech, Merck and Samus, and Pfizer.

Carole Ho: is employed by Denali Pharmaceuticals.

Robert Paul: is employed by Alector.

Rodney Pearlman: employed by The Bluefield Project

Margaret Sutherland is employed by the Chan Zuckerberg Initiative

Ajay Verma is employed by United Neuroscience.

Stephen Arneric: has nothing to disclose

Brian Alexander: has nothing to disclose

Bradford C. Dickerson: receives research support from NIH and royalties from Oxford University Press and Cambridge University; consults for Biogen, Merck, Lilly, Wave Lifesciences and Arkuda; and is paid by Elsevier for editorial activity.

Earl Ray Dorsey: has received honoraria for speaking at American Academy of Neurology courses, American Neurological Association, and University of Michigan; received compensation for consulting services from 23andMe, Abbott, Abbvie, American Well, Biogen, Clintrex, DeciBio, Denali Therapeutics, GlaxoSmithKline, Grand Rounds, Karger, Lundbeck, MC10, MedAvante, Medical-legal services, Mednick Associates, National Institute of Neurological Disorders and Stroke, Olson Research Group, Optio, Prilenia, Putnam Associates, Roche, Sanofi, Shire, Sunovion Pharma, Teva, UCB and Voyager Therapeutics; research support from Abbvie, Acadia Pharmaceuticals, AMC Health, Biosensics, Burroughs Wellcome Fund, Davis Phinney Foundation, Duke University, Food and Drug Administration, GlaxoSmithKline, Greater Rochester Health Foundation, Huntington Study Group, Michael J. Fox Foundation, National Institutes of Health/National Institute of Neurological Disorders and Stroke, National Science Foundation, Nuredis Pharmaceuticals, Patient-Centered Outcomes Research Institute, Pfizer, Prana Biotechnology, Raptor Pharmaceuticals, Roche, Safra Foundation, Teva Pharmaceuticals, University of California Irvine; editorial services for Karger Publications; and ownership interests with Blackfynn (data integration company) and Grand Rounds (second opinion service).

Murray Grossman: receives grant support from NIH, Avid and Piramal; participates in clinical trials sponsored by Biogen, TauRx, and Alector; serves as a consultant to Bracco and UCB; and serves on the Editorial Board of Neurology.

Edward Huey: has served as an investigator for clinical trials sponsored by AstraZeneca, Eli Lilly, and Roche / Genentech. He receives research support from Canadian Institutes of Health Research and the Alzheimer Society of British Columbia

Michael C. Irizarry is a full time employee of Eisai, Inc.

William J. Marks: is a full time employee of Verily, Inc.

Mario Masellis: has no relevant disclosures

Frances McFarland: received payment from Association for Frontotemporal Degeneration for writing support.

Debra Niehoff: employed by the Association for Frontotemporal Degeneration

Chiadikaobi Onyike: receives research funding from the NIH, the CIHR, and Biogen, Inc. He is also supported by the Jane Tanger Black Fund for Young-Onset Dementias, the Nancy H. Hall Fund for Geriatric Psychiatry, and a gift from Joseph Trovato.

Sabrina Paganoni: has received research funding from the Salah Foundation, the ALS Association, ALS Finding a Cure, the American Academy of Neurology, and Amylyx

Michael A Panzara is a fulltime employee of Wave Life Sciences.

Kenneth Rockwood: is President and Chief Science Officer of DGI Clinical, which in the last five years has contracts with pharma and device manufacturers (Baxter, Baxalta, Shire, Hollister, Nutricia, Roche, Otsuka) on individualized outcome measurement. In 2017 he attended an advisory board meeting with Lundbeck. Otherwise any personal fees are for invited guest lectures and academic symposia, received directly from event organizers, chiefly for presentations on frailty. He is Associate Director of the Canadian Consortium on Neurodegeneration in Aging, which is funded by the Canadian Institutes of Health Research, and with additional funding from the Alzheimer Society of Canada and several other charities, as well as, in its first phase (2013-2018), from Pfizer Canada and Sanofi Canada. He receives career support from the Dalhousie Medical Research Foundation as the Kathryn Allen Weldon Professor of Alzheimer Research, and research support from the Canadian Institutes of Health Research, the Nova Scotia Health Research Foundation, the Capital Health Research Fund and the Fountain Family Innovation Fund of the Nova Scotia Health Authority Foundation.

Jonathan Rohrer is supported by an MRC Clinician Scientist Fellowship (MR/M008525/1) and has received funding from the NIHR Rare Disease Translational Research Collaboration (BRC149/NS/MH). He has served on a Medical Advisory Board for Alector, Ionis and Wave Life Sciences.

Howard J Rosen: has received research support from Biogen Pharmaceuticals, has consulting agreements with Wave Neuroscience and Ionis Pharmaceuticals, and receives research support from NIH.

Robert Schuck: No conflicts of interest to disclose. This article reflects the views of the author and should not be construed to represent the FDA’s views or policies.

Holly D. Soares: is employed by AbbVie Pharmaceuticals and holds AbbVie stock shares.

Nadine Tatton: employed by the Association for Frontotemporal Degeneration

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Jicha GA, Nelson PT. Management of frontotemporal dementia: targeting symptom management in such a heterogeneous disease requires a wide range of therapeutic options. Neurodegenerative disease management. 2011;1:141–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Boxer AL, Gold M, Huey E, Gao FB, Burton EA, Chow T, et al. Frontotemporal degeneration, the next therapeutic frontier: molecules and animal models for frontotemporal degeneration drug development. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2013;9:176–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Tsai RM, Boxer AL. Therapy and clinical trials in frontotemporal dementia: past, present, and future. Journal of neurochemistry. 2016;138 Suppl 1:211–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Hodges JR, Piguet O. Progress and Challenges in Frontotemporal Dementia Research: A 20-Year Review. Journal of Alzheimer's disease : JAD. 2018;62:1467–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].O'Connor CM, Clemson L, Hornberger M, Leyton CE, Hodges JR, Piguet O, et al. Longitudinal change in everyday function and behavioral symptoms in frontotemporal dementia. Neurology Clinical practice. 2016;6:419–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Savage SA, Piguet O, Hodges JR. Cognitive intervention in semantic dementia: maintaining words over time. Alzheimer disease and associated disorders. 2015;29:55–62. [DOI] [PubMed] [Google Scholar]
  • [7].Henry ML, Hubbard HI, Grasso SM, Mandelli ML, Wilson SM, Sathishkumar MT, et al. Retraining speech production and fluency in non-fluent/agrammatic primary progressive aphasia. Brain : a journal of neurology. 2018;141:1799–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Finger EC. Frontotemporal Dementias. Continuum (Minneapolis, Minn). 2016;22:464–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Boxer AL, Gold M, Huey E, Hu WT, Rosen H, Kramer J, et al. The advantages of frontotemporal degeneration drug development (part 2 of frontotemporal degeneration: the next therapeutic frontier). Alzheimer's & dementia : the journal of the Alzheimer's Association. 2013;9:189–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Coyle-Gilchrist IT, Dick KM, Patterson K, Vazquez Rodriquez P, Wehmann E, Wilcox A, et al. Prevalence, characteristics, and survival of frontotemporal lobar degeneration syndromes. Neurology. 2016;86:1736–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Knopman DS, Roberts RO. Estimating the number of persons with frontotemporal lobar degeneration in the US population. Journal of molecular neuroscience : MN. 2011;45:330–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Onyike CU, Diehl-Schmid J. The epidemiology of frontotemporal dementia. International review of psychiatry (Abingdon, England). 2013;25:130–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Tsai RM, Boxer AL. Clinical trials: past, current, and future for atypical Parkinsonian syndromes. Seminars in neurology. 2014;34:225–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Boxer AL, Knopman DS, Kaufer DI, Grossman M, Onyike C, Graf-Radford N, et al. Memantine in patients with frontotemporal lobar degeneration: a multicentre, randomised, double-blind, placebo-controlled trial. Lancet neurology. 2013;12:149–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Boxer AL, Lang AE, Grossman M, Knopman DS, Miller BL, Schneider LS, et al. Davunetide in patients with progressive supranuclear palsy: A randomised, double-blind, placebo-controlled phase 2/3 trial. The Lancet Neurology. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Boxer AL, Yu JT, Golbe LI, Litvan I, Lang AE, Hoglinger GU. Advances in progressive supranuclear palsy: new diagnostic criteria, biomarkers, and therapeutic approaches. Lancet neurology. 2017;16:552–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Desmarais P, Rohrer JD, Nguyen QD, Herrmann N, Stuss DT, Lang AE, et al. Therapeutic trial design for frontotemporal dementia and related disorders. Journal of neurology, neurosurgery, and psychiatry. 2018. [DOI] [PubMed] [Google Scholar]
  • [18].Reiman EM, Langbaum JB, Tariot PN, Lopera F, Bateman RJ, Morris JC, et al. CAP--advancing the evaluation of preclinical Alzheimer disease treatments. Nature reviews Neurology. 2016;12:56–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Hoglinger GU, Respondek G, Stamelou M, Kurz C, Josephs KA, Lang AE, et al. Clinical diagnosis of progressive supranuclear palsy: The movement disorder society criteria. Movement disorders : official journal of the Movement Disorder Society. 2017;32:853–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Santos-Santos MA, Mandelli ML, Binney RJ, Ogar J, Wilson SM, Henry ML, et al. Features of Patients With Nonfluent/Agrammatic Primary Progressive Aphasia With Underlying Progressive Supranuclear Palsy Pathology or Corticobasal Degeneration. JAMA neurology. 2016;73:733–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Wood MJA, Talbot K, Bowerman M. Spinal muscular atrophy: antisense oligonucleotide therapy opens the door to an integrated therapeutic landscape. Human molecular genetics. 2017;26:R151–r9. [DOI] [PubMed] [Google Scholar]
  • [22].Iwamoto N, Butler DCD, Svrzikapa N, Mohapatra S, Zlatev I, Sah DWY, et al. Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides. Nature biotechnology. 2017;35:845–51. [DOI] [PubMed] [Google Scholar]
  • [23].Roberson ED, Hesse JH, Rose KD, Slama H, Johnson JK, Yaffe K, et al. Frontotemporal dementia progresses to death faster than Alzheimer disease. Neurology. 2005;65:719–25. [DOI] [PubMed] [Google Scholar]
  • [24].Mills SM, Mallmann J, Santacruz AM, Fuqua A, Carril M, Aisen PS, et al. Preclinical trials in autosomal dominant AD: implementation of the DIAN-TU trial. Revue neurologique. 2013;169:737–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Bateman RJ, Benzinger TL, Berry S, Clifford DB, Duggan C, Fagan AM, et al. The DIAN-TU Next Generation Alzheimer's prevention trial: Adaptive design and disease progression model. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2017;13:8–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Corriveau RA, Koroshetz WJ, Gladman JT, Jeon S, Babcock D, Bennett DA, et al. Alzheimer's Disease-Related Dementias Summit 2016: National research priorities. Neurology. 2017;89:2381–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Millenaar J, Hvidsten L, de Vugt ME, Engedal K, Selbaek G, Wyller TB, et al. Determinants of quality of life in young onset dementia - results from a European multicenter assessment. Aging & mental health. 2017;21:24–30. [DOI] [PubMed] [Google Scholar]
  • [28].Wu YT, Clare L, Hindle JV, Nelis SM, Martyr A, Matthews FE. Dementia subtype and living well: results from the Improving the experience of Dementia and Enhancing Active Life (IDEAL) study. BMC medicine. 2018;16:140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Mutsaerts H, Petr J, Thomas DL, De Vita E, Cash DM, van Osch MJP, et al. Comparison of arterial spin labeling registration strategies in the multi-center GENetic frontotemporal dementia initiative (GENFI). Journal of magnetic resonance imaging : JMRI. 2018;47:131–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Rohrer JD, Isaacs AM, Mizielinska S, Mead S, Lashley T, Wray S, et al. C9orf72 expansions in frontotemporal dementia and amyotrophic lateral sclerosis. Lancet neurology. 2015;14:291–301. [DOI] [PubMed] [Google Scholar]
  • [31].Bang J, Spina S, Miller BL. Frontotemporal dementia. Lancet. 2015;386:1672–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Bickart KC, Brickhouse M, Negreira A, Sapolsky D, Barrett LF, Dickerson BC. Atrophy in distinct corticolimbic networks in frontotemporal dementia relates to social impairments measured using the Social Impairment Rating Scale. Journal of Neurology, Neurosurgery & Psychiatry. 2013;85:438–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Kertesz A, McMonagle P, Blair M, Davidson W, Munoz DG. The evolution and pathology of frontotemporal dementia. Brain : a journal of neurology. 2005;128:1996–2005. [DOI] [PubMed] [Google Scholar]
  • [34].Kansal K, Mareddy M, Sloane KL, Minc AA, Rabins PV, McGready JB, et al. Survival in Frontotemporal Dementia Phenotypes: A Meta-Analysis. Dementia and geriatric cognitive disorders. 2016;41:109–22. [DOI] [PubMed] [Google Scholar]
  • [35].Brodtmann A, Cowie T, McLean C, Darby D. Phenocopy or variant: a longitudinal study of very slowly progressive frontotemporal dementia. BMJ case reports. 2013;2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Gomez-Tortosa E, Gallego J, Guerrero-Lopez R, Marcos A, Gil-Neciga E, Sainz MJ, et al. C9ORF72 hexanucleotide expansions of 20-22 repeats are associated with frontotemporal deterioration. Neurology. 2013;80:366–70. [DOI] [PubMed] [Google Scholar]
  • [37].Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain : a journal of neurology. 2011;134:2456–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Ducharme S, Dickerson BC. The neuropsychiatric examination of the young-onset dementias. The Psychiatric clinics of North America. 2015;38:249–64. [DOI] [PubMed] [Google Scholar]
  • [39].Cash DM, Bocchetta M, Thomas DL, Dick KM, van Swieten JC, Borroni B, et al. Patterns of gray matter atrophy in genetic frontotemporal dementia: results from the GENFI study. Neurobiology of aging. 2018;62:191–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Lee SE, Sias AC, Kosik EL, Flagan TM, Deng J, Chu SA, et al. Thalamo-cortical network hyperconnectivity in preclinical progranulin mutation carriers. NeuroImage Clinical. 2019;22:101751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Jacova C, Hsiung GY, Tawankanjanachot I, Dinelle K, McCormick S, Gonzalez M, et al. Anterior brain glucose hypometabolism predates dementia in progranulin mutation carriers. Neurology. 2013;81:1322–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Binney RJ, Pankov A, Marx G, He X, McKenna F, Staffaroni AM, et al. Data-driven regions of interest for longitudinal change in three variants of frontotemporal lobar degeneration. Brain and behavior. 2017;7:e00675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Ranasinghe KG, Rankin KP, Pressman PS, Perry DC, Lobach IV, Seeley WW, et al. Distinct Subtypes of Behavioral Variant Frontotemporal Dementia Based on Patterns of Network Degeneration. JAMA neurology. 2016;73:1078–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Perry DC, Brown JA, Possin KL, Datta S, Trujillo A, Radke A, et al. Clinicopathological correlations in behavioural variant frontotemporal dementia. Brain : a journal of neurology. 2017;140:3329–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Young AL, Marinescu RV, Oxtoby NP, Bocchetta M, Yong K, Firth NC, et al. Uncovering the heterogeneity and temporal complexity of neurodegenerative diseases with Subtype and Stage Inference. Nature communications. 2018;9:4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Staffaroni A, Cobigo Y, Goh S, Kornak J, Bajorek L, Chiang K, et al. Individualized Atrophy Scores Predict Dementia Onset in Familial Frontotemporal Lobar Degeneration. Alzheimer's and Dementia. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Rohrer JD, Rosen HJ. Neuroimaging in frontotemporal dementia. International review of psychiatry (Abingdon, England). 2013;25:221–9. [DOI] [PubMed] [Google Scholar]
  • [48].Knopman DS, Kramer JH, Boeve BF, Caselli RJ, Graff-Radford NR, Mendez MF, et al. Development of methodology for conducting clinical trials in frontotemporal lobar degeneration. Brain : a journal of neurology. 2008;131:2957–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Meeter LH, Kaat LD, Rohrer JD, van Swieten JC. Imaging and fluid biomarkers in frontotemporal dementia. Nature reviews Neurology. 2017;13:406–19. [DOI] [PubMed] [Google Scholar]
  • [50].Rojas JC, Karydas A, Bang J, Tsai RM, Blennow K, Liman V, et al. Plasma neurofilament light chain predicts progression in progressive supranuclear palsy. Annals of clinical and translational neurology. 2016;3:216–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Borroni B, Benussi A, Premi E, Alberici A, Marcello E, Gardoni F, et al. Biological, Neuroimaging, and Neurophysiological Markers in Frontotemporal Dementia: Three Faces of the Same Coin. Journal of Alzheimer's Disease. 2018;62:1113–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Chen Q, Boeve B, Senjem M, Tosakulwong N, Lesnick T, Przybelski S, et al. Rates of Lobar Atrophy in Asymptomatic MAPT Mutation Carriers. Azheimer's and Dementia. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Magdalinou NK, Paterson RW, Schott JM, Fox NC, Mummery C, Blennow K, et al. A panel of nine cerebrospinal fluid biomarkers may identify patients with atypical parkinsonian syndromes. Journal of neurology, neurosurgery, and psychiatry. 2015;86:1240–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Scherling CS, Hall T, Berisha F, Klepac K, Karydas A, Coppola G, et al. Cerebrospinal fluid neurofilament concentration reflects disease severity in frontotemporal degeneration. Annals of neurology. 2014;75:116–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Landqvist Waldo M, Frizell Santillo A, Passant U, Zetterberg H, Rosengren L, Nilsson C, et al. Cerebrospinal fluid neurofilament light chain protein levels in subtypes of frontotemporal dementia. BMC neurology. 2013;13:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Benatar M, Wuu J, Andersen PM, Lombardi V, Malaspina A. Neurofilament light: A candidate biomarker of presymptomatic amyotrophic lateral sclerosis and phenoconversion. Annals of neurology. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Rostgaard N, Roos P, Portelius E, Blennow K, Zetterberg H, Simonsen AH, et al. CSF neurofilament light concentration is increased in presymptomatic CHMP2B mutation carriers. Neurology. 2018;90:e157–e63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Ljubenkov PA, Staffaroni AM, Rojas JC, Allen IE, Wang P, Heuer H, et al. Cerebrospinal fluid biomarkers predict frontotemporal dementia trajectory. Annals of clinical and translational neurology. 2018;5:1250–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Rohrer JD, Woollacott IO, Dick KM, Brotherhood E, Gordon E, Fellows A, et al. Serum neurofilament light chain protein is a measure of disease intensity in frontotemporal dementia. Neurology. 2016;87:1329–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Meeter LH, Dopper EG, Jiskoot LC, Sanchez-Valle R, Graff C, Benussi L, et al. Neurofilament light chain: a biomarker for genetic frontotemporal dementia. Annals of clinical and translational neurology. 2016;3:623–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Skillback T, Mattsson N, Blennow K, Zetterberg H. Cerebrospinal fluid neurofilament light concentration in motor neuron disease and frontotemporal dementia predicts survival. Amyotrophic lateral sclerosis & frontotemporal degeneration. 2017;18:397–403. [DOI] [PubMed] [Google Scholar]
  • [62].Gaiani A, Martinelli I, Bello L, Querin G, Puthenparampil M, Ruggero S, et al. Diagnostic and Prognostic Biomarkers in Amyotrophic Lateral Sclerosis: Neurofilament Light Chain Levels in Definite Subtypes of Disease. JAMA neurology. 2017;74:525–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Hampel H, O'Bryant SE, Molinuevo JL, Zetterberg H, Masters CL, Lista S, et al. Blood-based biomarkers for Alzheimer disease: mapping the road to the clinic. Nature reviews Neurology. 2018;14:639–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].RW Paterson, Slattery CF, Poole T, Nicholas JM, Magdalinou NK, Toombs J, et al. Cerebrospinal fluid in the differential diagnosis of Alzheimer's disease: clinical utility of an extended panel of biomarkers in a specialist cognitive clinic. Alzheimer's research & therapy. 2018;10:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Jack CR Jr., Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2018;14:535–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].FDA. Early Alzheimer's Disease: Developing Drugs for Treatment Guidance for Industry. 2018https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM596728.pdf. [DOI] [PMC free article] [PubMed]
  • [67].FDA US. 2018. p. Table of Surrogate Endpoints That Were the Basis of Drug Approval or Licensure. https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/ucm613636.htm
  • [68].Devenney EM, Ahmed RM, Halliday G, Piguet O, Kiernan MC, Hodges JR. Psychiatric disorders in C9orf72 kindreds: Study of 1,414 family members. Neurology. 2018;91:e1498–e507. [DOI] [PubMed] [Google Scholar]
  • [69].DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72:245–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Whitwell JL, Jack CR Jr., Senjem ML, Parisi JE, Boeve BF, Knopman DS, et al. MRI correlates of protein deposition and disease severity in postmortem frontotemporal lobar degeneration. Neuro-degenerative diseases. 2009;6:106–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Lansdall CJ, Coyle-Gilchrist ITS, Jones PS, Vazquez Rodriguez P, Wilcox A, Wehmann E, et al. White matter change with apathy and impulsivity in frontotemporal lobar degeneration syndromes. Neurology. 2018;90:e1066–e76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Strong MJ, Abrahams S, Goldstein LH, Woolley S, McLaughlin P, Snowden J, et al. Amyotrophic lateral sclerosis - frontotemporal spectrum disorder (ALS-FTSD): Revised diagnostic criteria. Amyotrophic lateral sclerosis & frontotemporal degeneration. 2017;18:153–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].De Marchi F, Tondo G, Sarnelli MF, Corrado L, Solara V, D'Alfonso S, et al. A case of Progressive Non-Fluent Aphasia as onset of Amyotrophic Lateral Sclerosis with FrontoTemporal Dementia. The International journal of neuroscience. 2018:1–6. [DOI] [PubMed] [Google Scholar]
  • [74].Lindquist SG, Duno M, Batbayli M, Puschmann A, Braendgaard H, Mardosiene S, et al. Corticobasal and ataxia syndromes widen the spectrum of C9ORF72 hexanucleotide expansion disease. Clinical genetics. 2013;83:279–83. [DOI] [PubMed] [Google Scholar]
  • [75].Karch CM, Wen N, Fan CC, Yokoyama JS, Kouri N, Ross OA, et al. Selective Genetic Overlap Between Amyotrophic Lateral Sclerosis and Diseases of the Frontotemporal Dementia Spectrum. JAMA neurology. 2018;75:860–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Schuck RN, Woodcock J, Zineh I, Stein P, Jarow J, Temple R, et al. Considerations for Developing Targeted Therapies in Low-Frequency Molecular Subsets of a Disease. Clinical pharmacology and therapeutics. 2018;104:282–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Woodcock J, LaVange LM. Master Protocols to Study Multiple Therapies, Multiple Diseases, or Both. The New England journal of medicine. 2017;377:62–70. [DOI] [PubMed] [Google Scholar]
  • [78].Hyman DM, Piha-Paul SA, Won H, Rodon J, Saura C, Shapiro GI, et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature. 2018;554:189–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Hyman DM, Taylor BS, Baselga J. Implementing Genome-Driven Oncology. Cell. 2017;168:584–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Berry DA. Bayesian clinical trials. Nature reviews Drug discovery. 2006;5:27–36. [DOI] [PubMed] [Google Scholar]
  • [81].Rugo HS, Olopade OI, DeMichele A, Yau C, van 't Veer LJ, Buxton MB, et al. Adaptive Randomization of Veliparib-Carboplatin Treatment in Breast Cancer. The New England journal of medicine. 2016;375:23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Alexander BM, et al. INdividualized Screening trial of Innovative Glioblastoma Therapy (INSIGhT): A Bayesian adaptive platform trial (APT) to develop precision medicines for patients with GBM. JCO Precision Oncology. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Alexander BM, Ba S, Berger MS, Berry DA, Cavenee WK, Chang SM, et al. Adaptive Global Innovative Learning Environment for Glioblastoma: GBM AGILE. Clinical cancer research : an official journal of the American Association for Cancer Research. 2018;24:737–43. [DOI] [PubMed] [Google Scholar]
  • [84].Alexander BM, Cloughesy TF. Platform trials arrive on time for glioblastoma. Neuro-oncology. 2018;20:723–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Developing Personalized Clinical Outcome Assessments; April 5; The Richard J. Margolis Center for Strategic and International Studies; Washington, DC: 2017. p. 1–7. [Google Scholar]
  • [86].Cohen JA, Reingold SC, Polman CH, Wolinsky JS, International Advisory Committee on Clinical Trials in Multiple S. Disability outcome measures in multiple sclerosis clinical trials: current status and future prospects. Lancet Neurol. 2012;11:467–76. [DOI] [PubMed] [Google Scholar]
  • [87].Administration UFD. FDA Patient-Focused Drug Development Guidance Series for Enhancing the Incorporation of the Patient’s Voice in Medical Product Development and Regulatory Decision Making. In: CDER, editor.2018. [Google Scholar]
  • [88].Gaasterland CM, Jansen-van der Weide MC, Weinreich SS, van der Lee JH. A systematic review to investigate the measurement properties of goal attainment scaling, towards use in drug trials. BMC Med Res Methodol. 2016;16:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Northwestern University Hospital. Computer Adaptive Tests (CATs). 2018. [Google Scholar]
  • [90].Kiresuk TJ, Sherman RE. Goal attainment scaling: A general method for evaluating comprehensive community mental health programs. Community mental health journal. 1968;4:443–53. [DOI] [PubMed] [Google Scholar]
  • [91].Shabbir SH, Sanders AE. Clinical significance in dementia research: a review of the literature. American journal of Alzheimer's disease and other dementias. 2014;29:492–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Rockwood K, Fay S, Gorman M. The ADAS-cog and clinically meaningful change in the VISTA clinical trial of galantamine for Alzheimer's disease. International journal of geriatric psychiatry. 2010;25:191–201. [DOI] [PubMed] [Google Scholar]
  • [93].Rockwood K, Fay S, Song X, MacKnight C, Gorman M. Attainment of treatment goals by people with Alzheimer's disease receiving galantamine: a randomized controlled trial. CMAJ : Canadian Medical Association journal = journal de l'Association medicale canadienne. 2006;174:1099–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Rockwood K, Howlett SE, Hoffman D, Schindler R, Mitnitski A. Clinical meaningfulness of Alzheimer's Disease Assessment Scale-Cognitive subscale change in relation to goal attainment in patients on cholinesterase inhibitors. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2017;13:1098–106. [DOI] [PubMed] [Google Scholar]
  • [95].Wilz G, Weise L, Reiter C, Reder M, Machmer A, Soellner R. Intervention Helps Family Caregivers of People With Dementia Attain Own Therapy Goals. American journal of Alzheimer's disease and other dementias. 2018;33:301–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Scholzel-Dorenbos CJ, Meeuwsen EJ, Olde Rikkert MG. Integrating unmet needs into dementia health-related quality of life research and care: Introduction of the Hierarchy Model of Needs in Dementia. Aging & mental health. 2010;14:113–9. [DOI] [PubMed] [Google Scholar]
  • [97].Andrzejewski KL, Dowling AV, Stamler D, Felong TJ, Harris DA, Wong C, et al. Wearable Sensors in Huntington Disease: A Pilot Study. Journal of Huntington’s disease. 2016;5:199–206. [DOI] [PubMed] [Google Scholar]
  • [98].Espay AJ, Bonato P, Nahab FB, Maetzler W, Dean JM, Klucken J, et al. Technology in Parkinson's disease: Challenges and opportunities. Movement disorders : official journal of the Movement Disorder Society. 2016;31:1272–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Heldman DA, Harris DA, Felong T, Andrzejewski KL, Dorsey ER, Giuffrida JP, et al. Telehealth Management of Parkinson's Disease Using Wearable Sensors: An Exploratory Study. Digital biomarkers. 2017;1:43–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Hake AM, Dacks PA, Arneric SP. Concise informed consent to increase data and biospecimen access may accelerate innovative Alzheimer's disease treatments. Alzheimer's & dementia (New York, N Y). 2017;3:536–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Neville J, Kopko S, Romero K, Corrigan B, Stafford B, LeRoy E, et al. Accelerating drug development for Alzheimer's disease through the use of data standards. Alzheimer's & dementia (New York, N Y). 2017;3:273–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Arneric SP, Batrla-Utermann R, Beckett L, Bittner T, Blennow K, Carter L, et al. Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: A View of the Regulatory Science Qualification Landscape from the Coalition Against Major Diseases CSF Biomarker Team. Journal of Alzheimer's disease : JAD. 2017;55:19–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Albert D, Belsky DW, Crowley DM, Latendresse SJ, Aliev F, Riley B, et al. Can Genetics Predict Response to Complex Behavioral Interventions? Evidence from a Genetic Analysis of the Fast Track Randomized Control Trial. Journal of policy analysis and management : [the journal of the Association for Public Policy Analysis and Management]. 2015;34:497–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Tishchenko I, Riveros C, Moscato P. Alzheimer's disease patient groups derived from a multivariate analysis of cognitive test outcomes in the Coalition Against Major Diseases dataset. Future science OA. 2016;2:Fso140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Neu SC, Pa J, Kukull W, Beekly D, Kuzma A, Gangadharan P, et al. Apolipoprotein E Genotype and Sex Risk Factors for Alzheimer Disease: A Meta-analysis. JAMA neurology. 2017;74:1178–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Tang M, Gao C, Goutman SA, Kalinin A, Mukherjee B, Guan Y, et al. Model-Based and Model-Free Techniques for Amyotrophic Lateral Sclerosis Diagnostic Prediction and Patient Clustering. Neuroinformatics. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Dorsey ER, Venuto C, Venkataraman V, Harris DA, Kieburtz K. Novel methods and technologies for 21st-century clinical trials: a review. JAMA neurology. 2015;72:582–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Capell A, Liebscher S, Fellerer K, Brouwers N, Willem M, Lammich S, et al. Rescue of progranulin deficiency associated with frontotemporal lobar degeneration by alkalizing reagents and inhibition of vacuolar ATPase. J Neurosci. 2011;31:1885–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Sha SJ, Miller ZA, Min SW, Zhou Y, Brown J, Mitic LL, et al. An 8-week, open-label, dose-finding study of nimodipine for the treatment of progranulin insufficiency from GRN gene mutations. Alzheimer's & dementia (New York, N Y). 2017;3:507–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Cenik B, Sephton CF, Dewey CM, Xian X, Wei S, Yu K, et al. Suberoylanilide Hydroxamic Acid (Vorinostat) Up-regulates Progranulin Transcription. Journal of Biological Chemistry. 2011;286:16101–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Arrant AE, Filiano AJ, Unger DE, Young AH, Roberson ED. Restoring neuronal progranulin reverses deficits in a mouse model of frontotemporal dementia. Brain : a journal of neurology. 2017;140:1447–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Arrant AE, Onyilo VC, Unger DE, Roberson ED. Progranulin Gene Therapy Improves Lysosomal Dysfunction and Microglial Pathology Associated with Frontotemporal Dementia and Neuronal Ceroid Lipofuscinosis. The Journal of Neuroscience. 2018;38:2341–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Ly CV, Miller TM. Emerging antisense oligonucleotide and viral therapies for amyotrophic lateral sclerosis. Curr Opin Neurol. 2018;31:648–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Gendron TF, Chew J, Stankowski JN, Hayes LR, Zhang YJ, Prudencio M, et al. Poly(GP) proteins are a useful pharmacodynamic marker for C9ORF72-associated amyotrophic lateral sclerosis. Sci Transl Med. 2017;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Min SW, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nature medicine. 2015;21:1154–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].West T, Hu Y, Verghese PB, Bateman RJ, Braunstein JB, Fogelman I, et al. Preclinical and Clinical Development of ABBV-8E12, a Humanized Anti-Tau Antibody, for Treatment of Alzheimer's Disease and Other Tauopathies. J Prev Alzheimers Dis. 2017;4:236–41. [DOI] [PubMed] [Google Scholar]
  • [117].Bright J, Hussain S, Dang V, Wright S, Cooper B, Byun T, et al. Human secreted tau increases amyloid-beta production. Neurobiology of aging. 2015;36:693–709. [DOI] [PubMed] [Google Scholar]
  • [118].Novak P, Schmidt R, Kontsekova E, Zilka N, Kovacech B, Skrabana R, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer's disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet neurology. 2017;16:123–34. [DOI] [PubMed] [Google Scholar]
  • [119].Courade JP, Angers R, Mairet-Coello G, Pacico N, Tyson K, Lightwood D, et al. Epitope determines efficacy of therapeutic anti-Tau antibodies in a functional assay with human Alzheimer Tau. Acta neuropathologica. 2018;136:729–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Wang X, Smith K, Pearson M, Hughes A, Cosden ML, Marcus J, et al. Early intervention of tau pathology prevents behavioral changes in the rTg4510 mouse model of tauopathy. PLoS One. 2018;13:e0195486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].DeVos SL, Miller RL, Schoch KM, Holmes BB, Kebodeaux CS, Wegener AJ, et al. Tau reduction prevents neuronal loss and reverses pathological tau deposition and seeding in mice with tauopathy. Sci Transl Med. 2017;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Lunetta C, Lizio A, Maestri E, Sansone VA, Mora G, Miller RG, et al. Serum C-Reactive Protein as a Prognostic Biomarker in Amyotrophic Lateral Sclerosis. JAMA Neurol. 2017;74:660–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Miller RG, Block G, Katz JS, Barohn RJ, Gopalakrishnan V, Cudkowicz M, et al. Randomized phase 2 trial of NP001-a novel immune regulator: Safety and early efficacy in ALS. Neurol Neuroimmunol Neuroinflamm. 2015;2:e100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Le Pichon CE, Meilandt WJ, Dominguez S, Solanoy H, Lin H, Ngu H, et al. Loss of dual leucine zipper kinase signaling is protective in animal models of neurodegenerative disease. Science translational medicine. 2017;9. [DOI] [PubMed] [Google Scholar]
  • [125].Finger EC, MacKinley J, Blair M, Oliver LD, Jesso S, Tartaglia MC, et al. Oxytocin for frontotemporal dementia: a randomized dose-finding study of safety and tolerability. Neurology. 2015;84:174–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Cotelli M, Manenti R, Petesi M, Brambilla M, Cosseddu M, Zanetti O, et al. Treatment of primary progressive aphasias by transcranial direct current stimulation combined with language training. Journal of Alzheimer's disease : JAD. 2014;39:799–808. [DOI] [PubMed] [Google Scholar]
  • [127].Tippett DC, Hillis AE, Tsapkini K. Treatment of Primary Progressive Aphasia. Current treatment options in neurology 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES