3686 PART 16 Genes, the Environment, and Disease
■ IMMUNODEFICIENCY DISORDERS: PROOF OF
PRINCIPLE FOR EX VIVO GENE TRANSFER
Early attempts to effect gene replacement into hematopoietic stem cells
(HSCs) were stymied by the relatively low transduction efficiency of
retroviral vectors, which require dividing target cells for integration.
Because HSCs are normally quiescent, they are a formidable transduction target. However, identification of cytokines that induced cell
division without promoting differentiation of stem cells, along with
technical improvements in the isolation and transduction of HSCs, led
to modest but real gains in transduction efficiency.
The first convincing therapeutic effect from gene transfer occurred
with X-linked severe combined immunodeficiency disease (SCID),
which results from mutations in the gene (IL2RG) encoding the γc subunit of cytokine receptors required for normal development of T and
natural killer (NK) cells (Chap. 351). Affected infants present in the
first few months of life with overwhelming infections and/or failure to
thrive. In this disorder, it was recognized that successfully transduced
cells, even if few in number, would have a proliferative advantage compared to nontransduced cells, which lack receptors for the cytokines
required for lymphocyte development and maturation. Isolation of
autologous CD34+ cells, followed by transduction with a retroviral vector encoding the γc subunit and transplantation of the gene-modified
autologous cells, led to complete reconstitution of the immune system,
including documented responses to standard childhood vaccinations,
clearing of infections, and remarkable gains in growth in most of the
treated children. However, among 20 children treated in the initial
trials, five eventually developed a syndrome similar to T-cell acute
lymphocytic leukemia, with splenomegaly, rising white blood cell
counts, and the emergence of a single clone of T cells. Molecular studies revealed that, in most of these children, the retroviral vector had
integrated within a gene, LMO-2 (LIM only-2), that encodes a component of a transcription factor complex involved in hematopoietic
development. The retroviral long terminal repeat acted as a promoter
to increase the expression of LMO-2, resulting in T-cell leukemia.
The X-linked SCID studies were a watershed event in the evolution
of gene therapy. They demonstrated conclusively that gene therapy
could cure disease, with dramatic and durable clinical results. However,
they also demonstrated that insertional mutagenesis leading to cancer
was more than a theoretical possibility (Table 470-4). As a result of
the experience in these trials, all protocols using integrating vectors in
hematopoietic cells must include a plan for monitoring sites of insertion and clonal proliferation for 15 years after infusion (although to
date, insertional mutagenesis has never been reported in over several
thousand patient-years of experience where the transduced cell is a
mature T cell rather than an HSC). Strategies to overcome this complication have included using a “suicide” gene cassette in the vector, so
that errant clones can be quickly ablated, or using “insulator” elements
in the cassette, which can limit the activation of genes surrounding
the insertion site. The occurrence of malignancy in the X-linked SCID
trials has fueled a transition to lentiviral vectors. These efficiently
TABLE 470-2 Currently Approved Gene and Cell Therapy Products in the United States and/or Europe
PRODUCT INDICATION AGE GROUP
YEAR FIRST
APPROVED
WHERE
APPROVED VECTOR TRANSGENE TARGET TISSUE
Strimvelis®a ADA-SCID Pediatric 2016 Europe Retroviral ADA (adenosine
deaminase)
Autologous
hematopoietic stem
cells (HSCs)
Kymriah®
Tisagenlecleucel
Relapsed or refractory (R/R)
B-cell acute lymphoblastic
leukemia (pediatric); R/R large
B-cell lymphoma (adult)
Pediatric and
adult, different
disease indications
2017 U.S. and Europe Lentiviral Chimeric antigen
receptor with
4-1BB signaling
domain
Autologous T cells
Yescarta®
Axicabtagene
ciloleucel
R/R large B-cell lymphomas Adult 2017 U.S. and Europe Retroviral Chimeric antigen
receptor with CD28
signaling domain
Autologous T cells
Luxturna®
Voretigene
neparvovec
Confirmed biallelic RPE65
mutation-associated retinal
dystrophy
Pediatric and adult 2017 U.S. and Europe AAV RPE65 (retinal
pigment epithelial
65-kDa protein)
Retinal pigment
epithelial cells
Zolgensma®
Onasemnogene
abeparvovec
Spinal muscular atrophy type
1 due to biallelic mutations in
the SMN1 gene
Pediatric <2 years
of age
2019 U.S. and Europe AAV SMN1 (survival
motor neuron 1)
Spinal motor
neurons
Zynteglo®
Betibeglogene
autotemcel
Transfusion-dependent β
thalassemia
Adult and pediatric
≥12 years of age
2019 Europe Lentiviral bA-T87Q-globin gene Autologous HSCs
Libmeldy®b Metachromatic
leukodystrophy due to biallelic
mutations in the arylsulfatase
A gene
Pediatric 2020 Europe Lentiviral ARSA
(arylsulfatase A)
Autologous HSCs
Tecartus®
Brexucabtagene
autoleucel
R/R mantle cell lymphoma Adult 2020 U.S. Retroviral Chimeric antigen
receptor with CD28
signaling
Autologous T cells
a
Autologous CD34+-enriched cell fraction that contains CD34+ cells transduced with retroviral vector that encodes for the human ADA cDNA sequence. b
Autologous CD34+
cells encoding arylsulfatase A.
Abbreviations: AAV, adeno-associated virus; ADA-SCID, adenosine deaminase severe combined immunodeficiency.
TABLE 470-3 Most Common Indications in Gene Therapy Trials
INDICATION NUMBER
Cancer 1688
Monogenic diseases 287
Infectious diseases 182
Source: Adapted from SL Ginn et al: Gene therapy clinical trials worldwide to 2017:
An update. J Gene Med 20:e3015, 2018.
TABLE 470-4 Potential Complications of Gene Therapy
Gene silencing—repression of promoter
Genotoxicity—complications arising from insertional mutagenesis
Phenotoxicity—complications arising from overexpression or ectopic expression
of the transgene
Immunotoxicity—harmful immune response to either the vector or transgene; or
a harmful immune response of the vector (e.g., CAR T cells)
Risks of horizontal transmission—shedding of infectious vector into environment
Risks of vertical transmission—germline transmission of donated DNA
Abbreviation: CAR, chimeric antigen receptor.
Gene- and Cell-Based Therapy in Clinical Medicine
3687CHAPTER 470
transduce nondividing target cells and are characterized by a different
pattern of integration into the genome that appears to be safer than
retroviral vectors. To date, there have been no reports of tumorigenesis
due to insertional events with lentiviral vectors; the field is thus gradually moving toward these to replace retroviral vectors for transduction
of cells other than T cells.
Gene therapy also produced clear success for another form of SCID,
that due to adenosine deaminase (ADA) deficiency (Chap. 351).
ADA-SCID is clinically similar to X-linked SCID, although it can be
treated by enzyme replacement therapy with a pegylated form of the
enzyme (PEG-ADA), which leads to immune reconstitution but not
always to normal T-cell counts. Enzyme replacement therapy is expensive (annual cost: $200,000–$300,000 [U.S. dollars]), and some patients
develop antibodies to PEG-ADA, reducing therapeutic efficacy. The
initial trials of gene therapy for ADA-SCID were unsuccessful, but
modifications of this protocol to include the use of HSCs rather than
T cells as the target for transduction; discontinuation of PEG-ADA
at the time of vector infusion, so that the transduced cells have a
proliferative advantage over the nontransduced cells; and the use of a
mild conditioning regimen to facilitate engraftment of the transduced
autologous cells led to success without the complications seen in the
X-linked SCID trials. This therapy (Strimvelis®) was approved in 2016
by the European Medicines Agency (Table 470-2). Recently, however,
a patient who received the treatment in 2016 has been diagnosed with
T-cell leukemia, and preliminary findings suggest an insertional event
related to the gene therapy as the cause. Thus, although the risk of
insertional mutagenesis appears to be much lower in the setting of
ADA-SCID (1 patient out of 36 treated in the clinical trials or with
the approved product), it is not absent. Despite this, ADA-SCID is
an example where gene therapy has changed therapeutic options for
patients. For those with a human leukocyte antigen (HLA)-identical
sibling, bone marrow transplantation is still the best treatment option,
but this includes only a minority of those affected. For those without
an HLA-identical match, gene therapy has comparable efficacy to PEGADA, does not require repetitive injections, and does not present the
risk of development of neutralizing antibodies to the bovine enzyme.
Continued observation of previously treated patients may change the risk/
benefit assessment and perhaps promote development of lentivirus-based
vectors for the disease.
■ TRANSFUSION-DEPENDENT THALASSEMIA:
EXTENSION OF PRINCIPLE
A goal of gene therapy is to provide therapeutic options for more
prevalent diseases such as thalassemia and sickle cell disease. These
red cell disorders are more challenging targets for gene therapy than
the immunodeficiencies for several reasons: first, in immunodeficiency disorders, the transduced cells have a survival advantage over
nontransduced cells, which is not the case in red cell disorders. Second, in order to achieve transfusion independence or freedom from
vaso-occlusive crises, one must achieve higher transduction efficiency
and engraftment of higher numbers of stem cells. Promising clinical
data exist now for both of these conditions, and the first product for
transfusion-dependent β thalassemia has been conditionally approved
in Europe (Table 470-2). Standard of care for transfusion-dependent β thalassemia (TDT) consists of lifelong regular red blood cell
transfusions, typically monthly, to support hemoglobin levels >9 g/
dL, coupled with an intensive regimen of iron chelation to minimize
iron overload to the liver, heart, and endocrine system (Chap. 98).
Allogeneic stem cell transplantation addresses the underlying cause
of the disease but carries risks of myeloablation, graft-versus-host
disease (GVHD), and graft rejection and thus is reserved primarily
for those with an HLA-matched sibling donor (<25% of patients). The
first approved gene therapy for β thalassemia consists of a lentiviral
vector driving expression of an antisickling variant of β-globin (the
same product is being developed for sickle cell disease), introduced
into autologous HSCs, which are then transplanted back into the
patient after myeloablation. Results of clinical trials showed that
15 of 19 patients with TDT due to genotypes other than β0
/β0
(the
most severe genotype of the disease) achieved durable transfusion
independence that was maintained through follow-up ranging up to
5 years. The remaining four patients had substantial reduction in the
transfusion requirement. Gene therapy with lentiviral transduction of
autologous cells thus dramatically simplifies the medical regimen for
these patients, since it eliminates the need for transfusion and iron
chelation and carries no risk of GVHD or graft rejection because it is
generated from the patient’s own cells. Similarly, since the transduced
cells are autologous, there is no requirement for an HLA-matched
donor, expanding the numbers of patients who can be treated. Safety
in the initial trials has been excellent, with most adverse events related
to the known risks of the myeloablative conditioning regimen. Studies
currently underway aim to achieve the higher levels of transduction,
expression, and engraftment needed for patients with β0
/β0
thalassemia
and those with sickle cell disease.
Encouraging results using a gene-editing approach provide another
therapeutic option for these patients. In recent trials, a CRISPR/Cas9
vector targeting the BCL11A gene, which normally represses γ-globin
(the fetal β-like globin), was introduced into autologous CD34+ cells of
patients with TDT or sickle cell disease. Results with up to 18 months
of follow-up show a gradual rise in fetal hemoglobin in the circulation
to levels that obviate the need for transfusion and, in sickle cell disease, the cessation of vaso-occlusive crises. The gene-editing approach
still requires myeloablation to ensure engraftment of the gene-edited
autologous cells. Studies designed to support licensing are underway;
currently, there are no approved gene-editing products.
■ NEURODEGENERATIVE DISEASES: BROADENING
OF PRINCIPLE
The SCID trials gave support to the hypothesis that gene transfer into
HSCs could be used to treat any disease for which allogeneic bone
marrow transplantation was therapeutic. Moreover, the use of genetically modified autologous cells carried the advantages noted above,
i.e., no risk of GVHD, guaranteed availability of a “donor” (unless the
disease itself damages the stem cell population of the patient), and
low likelihood of failure of engraftment. Investigators in Paris capitalized on this realization to conduct the first trial of lentiviral vector
transduction of HSCs for a neurodegenerative disorder, X-linked
adrenoleukodystrophy (ALD). The key to the mechanism of action is
that a subpopulation of the gene-modified cells give rise to myeloid
cells that cross the blood-brain barrier and engraft as central nervous
system (CNS)–resident microglia and perivascular CNS macrophages.
The transduced cells carry the gene encoding the missing protein, in
this case an adenosine triphosphate–binding cassette transporter. Following lentiviral transduction of autologous HSCs in young boys with
the disease, dramatic stabilization of disease occurred, demonstrating
that stem cell transduction could work for neurodegenerative as well as
immunologic disorders.
Investigators in Milan carried this observation one step further
to develop a treatment for another neurodegenerative disorder that
had previously responded poorly to bone marrow transplantation.
Metachromatic leukodystrophy is a lysosomal storage disorder caused
by mutations in the gene encoding arylsulfatase A (ARSA). The late
infantile form of the disease is characterized by progressive motor
and cognitive impairment and death within a few years of onset, due
to accumulation of the ARSA substrate sulfatide in oligodendrocytes,
microglia, and some neurons. Recognizing that endogenous levels
of production of ARSA were too low to provide cross-correction
by allogeneic transplant, a lentiviral vector was engineered to direct
supraphysiologic levels of ARSA expression in transduced cells. Transduction of autologous HSCs from children born with the disease, at
a point when they were still presymptomatic, has led to preservation
and continued acquisition of motor and cognitive milestones at time
periods as long as 8 years after treatment, with observation ongoing.
This product is approved in Europe for those with late infantile or
early juvenile forms of the disease (Table 470-2). These results illustrate
that the ability to engineer levels of expression can allow gene therapy
approaches to succeed where allogeneic bone marrow transplantation
cannot. It is likely that a similar approach will be used in other neurodegenerative conditions.
3688 PART 16 Genes, the Environment, and Disease
LONG-TERM EXPRESSION IN GENETIC
DISEASE: IN VIVO GENE TRANSFER WITH
RECOMBINANT ADENO-ASSOCIATED
VIRAL VECTORS
Recombinant adeno-associated viral (AAV) vectors have emerged as
attractive gene delivery vehicles for genetic disease. Engineered from a
small replication-defective DNA virus, they are devoid of viral coding
sequences and trigger very little immune response in experimental animals. They are capable of transducing nondividing target cells, and the
donated DNA is stabilized primarily in an episomal form, thus minimizing risks arising from insertional mutagenesis. Because the vector
has a tropism for certain long-lived cell types, such as skeletal muscle,
the CNS, and hepatocytes, long-term expression can be achieved even
in the absence of integration. Of note, because the donated DNA is
predominantly nonintegrated, long-term expression requires targeting
of nondividing or slowly dividing cells; otherwise, expression is lost as
cells divide.
■ FIRST LICENSED PRODUCT
These features of AAV were used to develop the first approved gene
therapy product in the Western world, an AAV vector conditionally
approved in Europe in 2012 for treatment of the autosomal recessive
disorder lipoprotein lipase (LPL) deficiency. This rare disorder (1–2
cases/million) is due to loss-of-function mutations in the gene encoding LPL, an enzyme normally produced in skeletal muscle and required
for the catabolism of triglyceride-rich lipoproteins and chylomicrons.
Affected individuals have lipemic serum and may have eruptive xanthomas, hepatosplenomegaly, and in some cases, recurrent bouts of
acute pancreatitis, and do not respond to treatment with statins. Clinical trials demonstrated the safety of intramuscular injection of AAVLPL and provided preliminary evidence supporting a reduction in
frequency of episodes of pancreatitis. The sponsor allowed the approval
to expire in 2017, without completing the postmarketing requirements,
but the initial approval, demonstrating a regulatory pathway for this
class of therapeutics, was a crucial catalyst for the current robust
activity in gene therapy, with ongoing trials in a range of diseases
including muscular dystrophies, Parkinson’s disease, Huntington’s disease, Pompe disease, hemophilia B and A, several forms of congenital
blindness, and a variety of other inherited conditions. Currently there
are two approved AAV gene therapies for genetic disease, one for a rare
form of congenital blindness and the other for the fatal pediatric neurodegenerative disease spinal muscular atrophy type 1 (Table 470-2).
■ RETINAL GENE THERAPY
The retina is an attractive target for AAV-mediated gene transfer: It
is a relatively immuneprivileged space, obviating problems related to
immune responses, and the photoreceptors, retinal ganglion cells, and
retinal pigment epithelial cells are long-lived postmitotic cells. Routes
of administration for these cell types—either intravitreal or by subretinal injection—involve standard procedures in ophthalmology. Given
the small space, doses required are relatively low, lessening the manufacturing burden. Finally, canine models of a number of inherited retinal dystrophies have been well characterized and faithfully reflect the
human disease. Work carried out in the 1990s had demonstrated that
the canine disease could be reversed, with durable restoration of vision,
by subretinal injection of an AAV vector in dogs with a mutation in the
gene encoding RPE65 (retinal pigment epithelial-associated 65-kDa
protein), an enzyme key to the visual cycle. Like the canine disease, the
human disease is characterized by early-onset visual impairment, with
most patients progressing to blindness over time. Phase 1 clinical trials
by multiple groups established the safety of subretinal injections of an
AAV vector expressing RPE65. A phase 3 trial, the first randomized
controlled trial in human gene therapy, demonstrated improvement in
multiple measures of retinal and visual function. Of note, and likely to
be a recurring theme as gene therapies address diseases for which there
are no existing treatments, successful clinical development required
the development and validation of a novel clinical endpoint, a mobility
test conducted at varying levels of environmental illumination, that
could measure improvements in functional vision. This product, the
first licensed AAV gene therapy product in the United States, is now
approved in both the United States and Europe (Table 470-2). Trials for
other inherited retinal degenerative disorders such as choroideremia
are underway, as are studies for certain complex acquired disorders
such as age-related macular degeneration, which affects several million
people worldwide. The neovascularization that occurs in age-related
macular degeneration can be inhibited by expression of vascular endothelial growth factor (VEGF) inhibitors such as angiostatin or through
the use of RNA interference (RNAi)–mediated knockdown of VEGF.
Early-phase trials of an AAV vector designed to achieve long-term
inhibition of the biologic effects of VEGF through a soluble VEGF
receptor, however, failed to provide convincing evidence of efficacy,
illustrating the challenges of developing genetic approaches for complex acquired disorders.
■ SPINAL MUSCULAR ATROPHY TYPE 1
Spinal muscular atrophy type 1 is the most common genetic cause of
death in infancy and affects ~1 in 11,000 births. The disease is caused
by autosomal recessive mutations in the SMN1 gene, encoding survival motor neuron 1; affected infants undergo degeneration and loss
of lower motor neurons, presenting as hypotonia, severe weakness,
and failure to sit without support. In a large natural history study
of untreated infants with the disease, by age 20 months, only 8% of
patients with the disease were alive and free of ventilator support. In
a phase 1 gene therapy study, intravenous infusion of an AAV vector
(one with tropism for the nervous system [AAV9]) expressing SMN1
showed survival without ventilator support in 100% of participants
(n = 15) at 20 months of age. A phase 3 trial was initiated, but the
treatment was approved in the United States based on the efficacy
data from the first 21 participants enrolled in that study, coupled with
the safety data from the ongoing phase 3 study and the completed
phase 1 study (Table 470-2). The major safety concern was the risk of
acute serious liver injury; because the vector is infused intravenously,
there is considerable biodistribution to the liver (and to the spinal motor
neurons, the therapeutic target). The approved dose of 1.1 × 1014 vg/kg
is quite high and results in marked elevation of liver transaminases
if untreated. The phase 1 study showed that this could be controlled
using a course of corticosteroids begun 1 day before the vector infusion
and continued for 30 days, with tapering at that point and carried out
with monitoring of liver transaminases. Patients receiving the approved
treatment are followed in a registry designed to assess effectiveness, longterm safety, and overall survival of patients with spinal muscular atrophy.
An alternative treatment, intrathecal administration of an antisense
oligonucleotide designed to increase expression of functional SMN from
a mostly nonfunctional SMN2 gene, was approved during the course of
the gene therapy studies, but the efficacy data in the clinical trial were less
robust, and the drug requires intrathecal administration every 4 months.
Clearly, the gene therapy approach is a disease-modifying one, for a
disease that previously lacked treatment. Additional follow-up in this
population should address questions of long-term safety and efficacy.
GENE THERAPY FOR CANCER
The majority of clinical gene transfer experience has been in subjects
with cancer. The intent has been to increase the precision of cancer
therapies and thereby make them less toxic and more effective. Most
approaches have either modified the tumor directly or altered the host’s
response to the malignancy to produce immune effector cells that are
precisely targeted to the tumor phenotype.
■ MODIFYING THE CANCER
Since cancer is an (acquired) genetic disorder, initial efforts were
directed at correcting the genetic deficits of the tumor or introducing
lethal genes. Two major and persisting obstacles, however, are the poor
biodistribution and transduction efficiency of all currently available
vectors and the heterogeneity and genetic instability of the tumor targets themselves, so that correction of single driver mutations does not
preclude the evolution of a resistant population.
Tumor Correction One widely used direct intratumoral approach
was adenoviral-mediated expression of the tumor suppressor p53,
Gene- and Cell-Based Therapy in Clinical Medicine
3689CHAPTER 470
which is mutated in many different cancers. Initial studies showed
some complete and partial responses in squamous cell carcinoma of the
head and neck, esophageal cancer, and non-small-cell lung cancer, but
as yet, there have been no successful product licensing studies for this
approach except in China.
Prodrug Metabolizing Genes Efforts to overcome the above
limitations have included the introduction of a prodrug or a suicide
gene that would increase sensitivity of tumor cells to cytotoxic drugs.
An early strategy was intratumoral injection of an adenoviral vector
expressing the thymidine kinase (TK) gene. Cells that take up and
express the TK gene can be killed after the administration of ganciclovir, which is phosphorylated to a toxic nucleoside by TK. The advantage of this approach is that the effects of transducing even a limited
number of tumor cells are amplified by the spread of active drug to
adjacent tumor cells. Although the approach continues to be examined
in aggressive brain tumors and locally recurrent prostate, breast, and
colon tumors, progress remains slow, and systemic benefits against
metastatic disease have not been established.
■ MODIFYING THE HOST
Recruiting the Immune System The successful use of monoclonal antibodies that produce antitumor activity by activating the
immune response has demonstrated the feasibility of manipulating the
immune system to recognize the abnormal pattern of antigen expression on tumor cells. Immune cells are capable of almost unlimited
expansion and persistence and can provide long-term tumor control.
They can also traffic to tumor sites irrespective of location and, in principle, have the potential to evolve with the changing pattern of tumor
cell phenotype and function. Antibodies targeting “checkpoint” molecules, particularly CTLA-4 and the PD-1/PD-L1 axis, which naturally
limit T-cell responses and maintain tolerance, have been particularly
successful (Chap. 73).
Vaccination This strategy promotes more efficient recognition of
tumor cells by the immune system, but the development of a therapeutic vaccine, as opposed to the preventative vaccines required
to combat infectious diseases, has proved to be a considerable challenge. Approaches have included direct injection of tumor- or tumor
antigen–derived RNA or DNA; transduction of tumor cells with
immune-enhancing genes encoding cytokines, chemokines, or costimulatory molecules; and the ex vivo manipulation of dendritic cells
to enhance the presentation of tumor antigens. A dendritic cell vaccine
for treatment of recurrent prostate cancer has received approval in the
United States, but its limited potency and high cost have constrained
commercial success.
Adoptive Cell Transfer Host immune cells such as T cells, NK
cells, and others can be modified to express new transgenic receptors intended to recognize tumor cells and their microenvironment
(Fig. 470-1). Retargeting may use a modification of the cells’ own receptor or a molecularly synthesized chimeric antigen receptor (CAR) that
is usually composed of the antigen recognition portion of an antibody
and the signaling components of the cell’s native antigen receptor along
with one or more additional signaling domains that boost T-cell activation. Both approaches have been successful, with significant responses
reported with native receptors targeted to melanoma and synovial
cell sarcoma and—most dramatically—with CARs targeted to CD19,
an antigen expressed at high levels on normal and many malignant
B cells, or B-cell maturation antigen (BCMA), an antigen expressed
at high levels in normal and multiple myeloma plasma cells. Infused
CAR T cells can expand many thousand fold in vivo, persist long term,
and have produced >80% complete response rates when targeting
intractable B-cell acute lymphoblastic leukemia; approximately half of
treated patients have remained in remission for many years afterward
without further cancer therapies. This approach has also been successful in adult patients with relapsed or chemotherapy-refractory B
cell–derived large-cell lymphoma, mantle cell lymphoma, and multiple
myeloma. Many responses are sustained long term, and several of these
CAR T-cell products have been approved by the U.S. Food and Drug
Administration (FDA), as well as be international regulatory agencies,
and adopted as standard of care. The general approach of CAR T cells
is under rapid development, including trials with CAR T cells targeting
different antigens for solid tumors and other hematologic malignancies
and CAR T cells with different molecular structures and different gene
transfer vectors. Remaining challenges in the field and application of
adoptive T-cell approaches include (1) the immune inhibitory microenvironment associated with most solid tumors; recent studies further
modify the T cells with countermeasures to tumor inhibitory signals;
(2) acute and severe (though rarely fatal) systemic inflammatory and
neurologic toxicities during the phase of T-cell expansion and tumor
killing, which typically require access to intensive care for clinical
management; (3) the off-target or on-target but off-tumor effects that
may damage normal host tissues (e.g., normal B cells following CD19
CAR therapy); and (4) the cost, time, and complexity of manufacture,
which is a particular problem when antigens unique to each tumor’s
individual mutations are targeted (neoantigens), rather than widely
shared tumor-associated antigens.
Nonimmunologic Modifications to Host Gene transfer can be
used to protect normal cells from the toxicities of chemotherapy and
thereby increase the therapeutic index of these drugs. The most extensively
studied approach has been to transduce hematopoietic cells with genes
encoding resistance to chemotherapeutic agents, including the multidrug
resistance gene MDRI or the gene encoding O6
-methylguanine DNA
methyltransferase (MGMT). Although such approaches reduce hematologic toxicity, cytotoxic dose escalation quickly reveals dose-limiting
toxicities to other organ systems. Chemotherapy resistance can also be
engineered into immune cells redirected to target cancer to enable combination treatments with cells and chemotherapy.
Finally, gene transfer can be used to inhibit the host angiogenesis
required for tumor support, for example, by constitutive expression of
inhibitors such as angiostatin and endostatin or the transfer of T cells
genetically modified to recognize antigens specific to newly forming
vasculature. These studies are in the early phase.
■ COMBINATION APPROACHES—MODIFICATION
OF HOST AND TUMOR BY VIROTHERAPY
Immuno-oncolytic Viruses These viruses are genetically modified to replicate in malignant but not normal cells. The replicating
CH2 VH
CL
CH
CAR T cells Native T cells
T cells
Antigenic
peptide
TCR complex
CAR
Spacer
CD3ζ
COSTIM
TM
TCR complex
γ ε ε δ ζ
γ ε ε δ ζ ζ
Monoclonal
antibody
Target cell TCR
β2
β
β
α
α
MHC I
VL
CH3
VL
VH
ζ ζ
FIGURE 470-1 T-cell receptors. A native T-cell receptor (TCR) recognizes processed
peptide antigens bound to major histocompatibility (MHC) molecules through its
αβ chains. Signaling then occurs through a multichain intracellular CD3 complex.
A chimeric antigen receptor (CAR) usually contains an extracellular receptor
component derived from the antigen binding portion (VH and VL
) of a monoclonal
antibody. This produces a receptor that can recognize either protein or nonprotein
antigens independent of the MHC. A transmembrane (TM) domain then connects
this receptor to the ζ chain of the CD3 complex derived from the native TCR. A
costimulation domain (COSTIM), such as CD28 or 4-1BB, is also present.
3690 PART 16 Genes, the Environment, and Disease
TABLE 470-5 Taking History from Subjects Enrolled in Gene Transfer
Studies
Elements of History for Subjects Enrolled in Gene Transfer Trials
1. What vector was administered? Is it predominantly integrating (retroviral,
lentiviral, herpesvirus [latency and reactivation]) or nonintegrating (plasmid,
adenoviral, adeno-associated viral)?
2. What was the route of administration of the vector?
3. What was the target tissue?
4. What gene was transferred in? A disease-related gene? A marker?
5. Were there any adverse events noted after gene transfer?
Screening Questions for Long-Term Follow-Up in Gene Transfer
Subjectsa
1. Has a new malignancy been diagnosed?
2. Has a new neurologic/ophthalmologic disorder, or exacerbation of a
preexisting disorder, been diagnosed?
3. Has a new autoimmune or rheumatologic disorder been diagnosed?
4. Has a new hematologic disorder been diagnosed?
a
Factors influencing long-term risk include integration of the vector into the
genome, vector persistence without integration, and transgene-specific effects.
“All disease begins in the gut.”
—Hippocrates
Nearly two and a half millennia after Hippocrates made this statement,
we are just coming to truly appreciate its profundity. Since the beginning of humankind, scholars have been investigating the underpinnings of disease with an almost singular focus on the human side of
the equation. Microbes were not recognized as an important cause of
disease until the inception of the “germ theory” in the late nineteenth
century. During the first century of medical microbiology, research
largely centered on the role of microbes as pathogens. Only recently has
there been a resurgence of interest in understanding how commensal
organisms—the bacteria, viruses, fungi, and Archaea that make up
the microbiota—impact human physiology. The idea that these microorganisms are vital to the well-being of humans has challenged our
traditional notions of “self.” Indeed, a human being can most accurately
be described as a holobiont: a complex assemblage of human cells and
microorganisms interacting in an elaborate pas de deux that drives
normal physiologic processes.
Aimed at a better understanding of this relationship, myriad studies during the past decade have begun to catalogue the microbiota at
various body sites and in a multitude of disease conditions. Diseases in
virtually every organ system have been associated with changes in the
microbiota. Indeed, the microbiota has been linked to intestinal disorders, disturbances in metabolic function, autoimmune diseases, and
psychiatric conditions and has been shown to influence susceptibility
to infection and the efficacy of pharmaceutical therapies. Knowledge
of the specific mechanism(s) underlying most of these microbe–disease
associations is lacking; it remains unclear whether the disease-associated
alterations in the microbiota represent mere biomarkers of disease, a
471 The Human Microbiome
Neeraj K. Surana, Dennis L. Kasper
vectors thus proliferate and spread within the tumor, facilitating eventual tumor clearance. However, physical limitations to viral spread,
including fibrosis, intermixed normal cells, basement membranes,
and necrotic areas within the tumor, may reduce clinical efficacy, and
their activity against metastatic disease has proved limited. Recently,
the FDA granted licensing approval to talimogene laherparepvec, an
oncolytic herpes virus containing the human granulocyte-macrophage
colony-stimulating factor gene, for treatment of melanoma. This
success has led to resurgent interest in combining the local tumor
destruction and tumor antigen release mediated directly by oncolytic
viruses with the recruitment of a systemic immune response mediated
by immunostimulatory genes contained within the oncolytic virus. In
principle, such immune-oncolytic viruses should produce responses in
both local and metastatic disease. Numerous novel viral agents are now
entering early-phase clinical testing.
■ OTHER APPROACHES
This chapter has focused on gene addition therapy, in which a normal
gene is transferred to a target tissue to drive expression of a gene product with therapeutic effects. Another powerful technique beginning
to yield dramatic clinical results is genome editing, which has the
potential to correct a mutation in situ, generating a wild-type copy
under the control of the endogenous regulatory signals. This approach
makes use of novel reagents including zinc finger nucleases, TALENs,
and CRISPR, which introduce breaks into the DNA near the site of the
mutation and then rely on a donated repair sequence and cellular mechanisms for repair to reconstitute a functioning gene. Another strategy
recently introduced into clinical trials is the use of small interfering
RNAs or short hairpin RNAs as transgenes to knock down expression
of deleterious genes (e.g., mutant huntingtin in Huntington’s disease).
SUMMARY
The power and versatility of gene therapy approaches are such that
there are few serious disease entities for which gene therapies are
not under development. The development of new classes of therapeutics typically takes two to three decades; monoclonal antibodies
and recombinant proteins are recent examples. Gene therapeutics,
which entered clinical testing in the early 1990s, traversed the same
time course. Examples of clinical success are now abundant, and gene
therapy approaches are likely to become increasingly important as a
therapeutic modality in the twenty-first century. A central question to
be addressed is the long-term safety of gene transfer, and regulatory
agencies have mandated a 15-year follow-up for subjects enrolled in
gene therapy trials (Table 470-5). Realization of the therapeutic benefits of modern molecular medicine will depend on continued progress
in gene transfer technology.
■ FURTHER READING
Al-Zaidy SA et al: AVXS-101 (onasemnogene abeparvovec) for
SMA1: Comparative study with a prospective natural history cohort.
J Neuromusc Dis 6:307, 2019.
Fesnak AD et al: Engineered T cells: The promise and challenges of
cancer immunotherapy. Nat Rev Cancer 16:566, 2016.
Fischer A, Hacein-Bey-Abina S: Gene therapy for severe combined
immunodeficiencies and beyond. J Exp Med 217:e20190607, 2020.
Frangoul H et al: CRISPR-Cas9 gene editing for sickle cell disease
and b-thalassemia. N Engl J Med 384:252, 2021.
High KA, Roncarolo MG: Gene therapy. N Engl J Med 381:455,
2019.
Hocquemiller M et al: Adeno-associated virus-based gene therapy
for CNS diseases. Hum Gene Ther 27:478, 2016.
June CH, Sadelain M: Chimeric antigen receptor therapy. N Engl J
Med 379:64, 2018.
Ramos CA et al: CAR-T cell therapy for lymphoma. Annu Rev Med
67:165, 2016.
Russell S et al: Efficacy and safety of voretigene neparvovec (AAV2-
hRPE65v2) in subjects with RPE65-mediated inherited retinal dystrophy: A randomised, controlled, open label phase 3 trial. Lancet
390:849, 2017.
Sessa M et al: Lentiviral haemopoietic stem-cell gene therapy in
early-onset metachromatic leukodystrophy: An ad-hoc analysis of a
non-randomised, open-label, phase 1/2 trial. Lancet 388:476, 2016.
Thompson AA et al: Gene therapy in patients with transfusiondependent β-thalassemia. N Engl J Med 378:1479, 2018.
The Human Microbiome
3691CHAPTER 471
diseases. Thus, although the field of microbiome research is sometimes
considered to have emerged over the last one or two decades, the basic
tenets—that the microbiota varies according to body site and clinical
characteristics, that microbes are critical for human health, and that
specific modulation of the microbiota may lead to improved clinical
outcomes—are far from new.
A PRIMER ON TAXONOMY
Given that microbiome-based studies have identified and compared
microbes at different levels of taxonomic resolution (Fig. 471-1), some
understanding of taxonomy is essential for better comprehension of
the implications of these studies. Of the ~100 bacterial phyla that
exist in nature, only five (Actinobacteria, Bacteroidetes, Firmicutes,
Fusobacteria, and Proteobacteria) are dominant members of the
human microbiome. Each of these phyla can be further categorized
into multiple classes, orders, families, genera, and species. Early studies
on the microbiota focused on changes in the relative abundance at the
phylum level between different groups (e.g., obese vs normal-weight
patients); however, these comparisons are at such a broad taxonomic
level that they often provide little or no biologic insight. As illustrated
in Fig. 471-1, drawing comparisons between organisms in two different bacterial phyla is analogous to comparing humans to sea stars: the
evolutionary distance between the two is tremendous. The limitations
of current bioinformatic tools require lumping together of taxonomically related strains and thus cloud the richness of microbial ecology.
Examining microbial profiles at the phylum, family, or even genus
level—as is often done at present—ignores the great heterogeneity
within different strains of the same bacterial species. The analytical
pipelines are just now beginning to enable strain-level comparisons,
and these improvements will likely facilitate our ongoing investigation
of host–commensal interactions.
THE MICROBIOTA AND HUMAN HEALTH
■ OVERVIEW OF THE HUMAN MICROBIOTA
The overwhelming majority of microbiota studies have focused on
stool, given that this sample type represents the most ecologically rich
anatomic site, is easy to obtain, and can readily be followed longitudinally in the same individual. A landmark study by the HMP sought to
define the “normal” microbiota throughout the entire body in healthy
Western adults. To this end, the microbial populations at 15–18 body
sites were characterized in 242 people. One striking finding was that
all samples from a given body region (e.g., skin) were more similar
to each other than they were to samples from a different body region
(e.g., stool), even in the same individual (Fig. 471-2A). In essence, the
effect of the anatomic site on microbial composition is far greater than
the effect of heterogeneity between individuals. That said, there was
a remarkable amount of interindividual variation at any given body
site (Fig. 471-2B). In stool, for example, the abundance of the phylum
Domain
Phylum
Kingdom
Class
Order
Family
Genus
Species
Staphylococcus
Staphylococcaceae
Bacillales
Bacilli
Firmicutes
Bacteria
Primate
Hominidae
Homo
Mammalia
Chordate
Animalia
Eukarya
S. aureus
S. epidermidis
S. lugdunensis
G. haemolysans
M. equipercicus
L. monocytogenes
B. anthracis
S. pneumoniae
E. faecalis
C. botulinum
C. difficile
E. rhusiopathiae
E. coli
B. fragilis
FIGURE 471-1 Juxtaposition of bacterial and human taxonomy highlights the evolutionary distance between different taxonomic levels. The listed species represent
exemplars that are members of the taxon to which they are connected but that are not contained within the next-lower-level taxon listed. For example, Clostridium botulinum,
Clostridium difficile, and Erysipelothrix rhusiopathiae are members of the phylum Firmicutes, but are in classes other than Bacilli. Similarly, starfish and humans are both
members of the kingdom Animalia, but they are in different phyla.
causal relationship, or a combination of the two. Although cause-andeffect relationships are still being elucidated for many diseases, it is
clear that humans coexist in an intricate relationship with commensal
organisms. This chapter explores in detail the nature of these host–
commensal interactions, focusing on how this information might be
translated into clinically meaningful interventions.
HISTORICAL PERSPECTIVE
Massive undertakings, such as the Human Microbiome Project (HMP)
sponsored by the National Institutes of Health and MetaHIT sponsored
by the European Commission, have catalogued all the bacteria present
at multiple body sites in people with and without disease. Coupled with
the confluence of advances in sequencing technologies (Chap. 121),
gnotobiotic animal availability, and microbial culture, significant progress has been made toward an understanding of the interplay between
the microbiota and human health. However, recent findings were foreshadowed by work done centuries ago.
The human microbiota was first explored in 1683 when Antony
van Leeuwenhoek described in a letter to the Royal Society of London
the “very little living animalcules, very prettily a-moving” that he had
observed in the plaque between his teeth. Leeuwenhoek went on to
perform the first comparative “microbiota” studies by assessing how
fecal and oral bacteria differ, how oral microbes change in the setting
of disease (e.g., alcoholism and tobacco use), and how microbial composition changes across the age spectrum (e.g., in young children vs
old men). He attempted—unsuccessfully—to eliminate these bacteria.
Although Leeuwenhoek was not taken seriously when he first reported
his findings, his studies laid the groundwork for what is now the field
of microbiome research, and investigators are still trying to answer
many of the same overarching questions that he raised more than three
centuries ago.
Although Leeuwenhoek first reported the existence of bacteria and
their association with humans at the end of the seventeenth century,
the significance of commensal bacteria was not realized until late in
the nineteenth century. In 1885, Pasteur suggested that animals could
not survive if they were “artificially and completely deprived of the
common microbes.” Although Pasteur’s preconceived ideas were proven
incorrect in 1912 by the advent of germ-free animals (animals raised
without exposure to any microorganisms), the underlying concept that
commensal organisms are critical to health has held up. Élie Metchnikoff
made another conceptual advance in this field by suggesting at the
beginning of the twentieth century that clinical outcomes could be
altered by the administration of specific beneficial organisms (probiotics). In particular, Metchnikoff believed that aging was caused by
toxic bacteria in the gut and that lactic acid–producing bacteria (e.g.,
Lactobacillus species) present in sour milk and yogurt could mitigate
against this process. The data behind this specific claim are still lacking, but recent discoveries offer continued hope that the microbiome
can be effectively harnessed to protect against and treat a variety of
3692 PART 16 Genes, the Environment, and Disease
Bacteroidetes ranged from ~10% in some individuals to >90% in others. Remarkably, even with person-to-person variability and differences
among body sites, the functional capacity of the microbiota—assessed
using metagenomic data to identify gene pathways—was quite similar
across different people and different body sites (Fig. 471-2C). This discrepancy between the substantial differences in microbial composition
and the little or no resulting change in the functional properties of the
microbiota reflects an important ecologic property of the microbiota:
the microbial communities at different body sites and in different
people assemble in such a way that all the core metabolic functions
are maintained. This finding also hints at the likely possibility of significant functional redundancy within the microbiota, with different
species executing the same biologic functions in different people and/
or at different anatomic sites.
While the HMP provided the first large-scale catalogue of the microbiome in multiple people and at many different body sites, the amount
of data generated by what, at the time, was by far the largest microbiome study has been dwarfed by subsequent studies. These more recent
studies have confirmed the HMP’s major tenets: the composition of
the microbiota differs by body site, there is tremendous interindividual
variation, and the microbial gene content is relatively conserved irrespective of the body site or individual. No microbial species are ubiquitous in all individuals and at all body sites, but some species are highly
prevalent at a given body site: in the HMP study, Staphylococcus epidermidis was present in 93% of nares samples and Escherichia coli in 61%
of stool samples. These findings highlight the remarkable personalization of the human microbiome. While the human genome is typically
>99.5% identical in different people, the microbiotas of two individuals
may not overlap at all. Although the “precision medicine” approach
currently focuses on teasing out how differences in the human genome
relate to different clinical end points, the human microbiome clearly
represents a critical component for consideration.
■ THE MICROBIOTA BY THE NUMBERS
It has long been known that the human-associated microbiota is numerically dense. Leeuwenhoek estimated that there were more “animals
living in the scrum on the teeth in man’s mouth than there are men in
a kingdom.” Specific enumeration of the components of the microbiota
has been challenging, in part because of its variability across time,
space (body region), and clinical conditions. Moreover, the majority
of human-associated microbes are not readily cultivable—a situation
that raises questions about the best methodology for such quantitation.
Initial back-of-the-envelope calculations performed in the 1970s suggested that there were roughly tenfold more bacteria in the body than
there were human cells. This rather astounding estimate suggested
that humans are really only ~10% “human” and that by far the greatest
part of the holobiont is represented by microbes. This stark numerical
discrepancy has prompted some to question “who parasitizes whom.”
However, a more recent estimate has suggested that there are “only”
~1.3 times more bacteria in the body than there are human cells and
thus that humans are ~56% “bacterial.” Of note, this more recent
study does not include the numbers of viruses (known to generally
be approximately tenfold more abundant than other microbes), fungi,
or Archaea. Given these additional microorganisms, the notion that
microbes constitute >90% of the cells present in a human body is
likely correct. These ratios are even starker when one considers the
genetic potential of human cells versus that of commensal organisms.
In contrast to the ~20,000 genes in the human genome, the estimated
Phyla
Firmicutes
Actinobacteria
Bacteroidetes
Proteobacteria
Fusobacteria
Tenericutes
Spirochaetes
Cyanobacteria
Verrucomicrobia
TM7
Metabolic pathways
Central carbohydrate metabolism
Cofactor and vitamin biosynthesis
Oligosaccharide and polyol transport system
Purine metabolism
ATP synthesis
Phosphate and amino acid transport system
Aminoacyl tRNA
Pyrimidine metabolism
Ribosome
Aromatic amino acid metabolism
B
C
Anterior nares RC Buccal mucosa Supragingival plaque Tongue dorsum Stool Posterior fornix PC2 (4.4%)
PC1 (13%)
Gastrointestinal
Urogenital
Skin
Nasal
Oral
A
FIGURE 471-2 The human microbiome exhibits significant taxonomic variability among body sites and between individuals while maintaining core metabolic pathways.
A. Principal coordinates (PC) plot showing variation among samples demonstrates that primary clustering is by body area, with the oral, gastrointestinal, skin, and urogenital
habitats separate; the nares habitat bridges oral and skin habitats. Each circle represents an individual sample. B, C. Vertical bars represent microbiome samples by
body habitat, with each bar within a given body site representing a different individual. Bars indicate relative abundances colored by microbial phyla (B) and metabolic
pathways (C). The legend on the right indicates the most abundant phyla/pathways. RC, retroauricular crease. (Reproduced with permission from Human Microbiome
Project Consortium: Structure, function and diversity of the healthy human microbiome. Nature 486:207, 2012.)
The Human Microbiome
3693CHAPTER 471
Supragingival plaque
GI/Stool
Skin
Vagina
Actinobacteria
Bacteroidetes
Fusobacteria
Proteobacteria
Firmicutes
Other
KEY
Buccal mucosa
Nares
FIGURE 471-3 Different anatomic sites harbor very different microbiomes. The figure indicates the relative
proportion of sequences determined at the taxonomic phylum level at six anatomic sites. (Data for stool, vagina,
nares, buccal mucosa, and supragingival plaque are from the Human Microbiome Project; data for the skin are
from EA Grice et al: Topographical and temporal diversity of the human skin microbiome. Science 324:1190,
2009.)
total number of genes in the microbiota (which
together constitute the microbiome)—i.e.,
>2,000,000—indicates that the human genome
contributes <1% to the total genetic potential
of the overall holobiont. Most microbiome
studies to date have focused almost exclusively
on the bacterial component; much remains to
be learned about the functional interplay of
bacteria, viruses, fungi, and Archaea and how
these other classes of microorganisms impact
human health.
In terms of overall diversity, >10,000 different bacterial species are present in the human
microbiota; the intestines alone contain >1000
species. At any given time, the body of any given
individual harbors 500–1000 bacterial species,
with 100–200 bacterial species in the gut alone.
If one considers different strains of the same
bacterial species, which may be functionally
different from one another, the diversity of the
microbiota is probably at least a magnitude
greater. Although marked diversity exists at the
strain and species level, only limited bacterial
phyla are generally found in the human microbiota at any given body site (Fig. 471-3).
■ INFLUENCES ON THE
MICROBIOTA
An individual’s specific microbial configuration
is dynamic and is quickly altered in response
to subtle changes in the microenvironments
in which the bacteria reside. On a day-to-day
basis, these changes usually reflect alterations in
the relative abundance of the various microbes.
However, some exposures have a greater effect
on the microbiota and can shift the microbial
population to a new equilibrium via the loss
of specific species and/or the acquisition of
others; this new microbial equilibrium can be
associated with either health or a disease state
(Fig. 471-4). Identification of the factors that influence the microbiota’s composition is critical to an understanding of what leads to and
controls intra- and interindividual variation. Moreover, an understanding of the influences on the microbiota will facilitate the design and
proper interpretation of microbiota studies. While it is clear that the
microbiota can be altered through these various mechanisms, it is not
yet clear whether these changes are biologically significant.
Genetics Studies of monozygotic and dizygotic twins have revealed
that host genetics have a small but statistically significant effect on the
microbiota’s composition. Notably, some taxa, such as Christensenella
species, are more heritable than others. A cross-sectional study of
>1000 healthy individuals who have distinct ancestral origins and a
relatively shared common environment confirmed a weak association
between host genetics and the microbiome but highlighted that environmental factors are more prominent modulators of the microbiome.
That said, the host’s genetic contribution to the microbiota, albeit
small, may be meaningful. Studies in mice have demonstrated that
genetic variation in the major histocompatibility complex, a specific set
of immune-related genes, leads to changes in the microbiota that alter
susceptibility to an autoimmune disease. These studies offer a proof
of concept for the notion that the genetic predisposition observed for
certain diseases may actually be mediated by indirect alterations in the
microbiota.
Age Burgeoning evidence now indicates that microbial exposure
may begin in utero: bacterial DNA from bacteria typically associated
with the oral microbiota has been identified in otherwise healthy
placentas, in amniotic fluid obtained at early stages of gestation, and
in meconium of term newborns. Although some controversy persists
about whether these results reflect contamination and/or the presence
of nonviable bacteria, they raise the possibility that human exposure to
the microbial world begins before birth. The delivery mode (vaginal vs
cesarean section) and the method of feeding (breast milk vs formula,
timing of solid food introduction) are major determinants of an infant’s
Unstable
Stable
Healthy state 1
Current microbial
state
Disease state
Healthy state 2
FIGURE 471-4 A stability landscape of the human microbial ecosystem. A stable
state, illustrated as a depression in the landscape, can be associated with either a
healthy state or a disease state. The topology of an individual’s landscape reflects
that person’s genetics, age, diet, medications, medical history, and lifestyle. The
position of the green ball represents the current microbial state. Clinical changes
(e.g., administration of antibiotics, development of disease) can influence both the
current state and the overall topology.
3694 PART 16 Genes, the Environment, and Disease
1
D Strachan: BMJ 299:1259, 1989.
early microbiota. After birth, the infant’s microbiota goes through a
stereotyped succession process; with increases in bacterial diversity
and functional capacity, the child’s microbiota resembles that of an
adult by the age of 2–3 years. Cross-sectional studies that have examined the microbiota across the entire age spectrum have revealed a
general stability of the fecal microbiota after 2–3 years of age; however,
the microbiota of the elderly (persons >80 years of age) demonstrates
notable differences from those of their younger counterparts, with
increases in Bacteroides and Eubacterium species and decreases in the
bacterial family Lachnospiraceae.
Diet Diet is a strong determinant of human health. The impact of
diet is mediated, in part, by its effects on the composition of the gut
microbiota. This makes intuitive sense, as the human diet provides
nutrients needed not only by our own cells but also by the microbes
living in the alimentary tract. In young children, this dietary influence
is marked by major shifts (e.g., a decrease in Bifidobacterium species) in
the intestinal microbiota that occur at weaning and with the introduction of solid food. In adults, long-term dietary patterns are associated
with relatively stable microbial compositions. However, drastic changes
in short-term macronutrient availability cause rapid (within 1 day)
and reproducible fluctuations in the fecal microbiota that reflect the
biologic processes needed to degrade and metabolize the nutrients in
the new diet. For example, vegetarian diets are associated with a microbiota that has an increased ability to metabolize plant polysaccharides
(e.g., Roseburia species, Eubacterium rectale, Ruminococcus bromii),
while animal-based diets result in an increased abundance of biletolerant organisms (e.g., Alistipes, Bilophila, and Bacteroides species).
At the completion of dietary interventions and the resumption of the
individual’s normal dietary pattern, the microbial communities revert
back to their previous states, probably because the individual resumes
his or her typical diet. Taken together, dietary studies confirm that
the microbiota is highly adaptable and varies in relation to changes in
the diet. Of note, virtually all of these studies have focused on how the
diet influences the fecal microbiota. It will be interesting to determine
whether dietary changes similarly influence the microbiota at nonintestinal sites.
Drugs Virtually all drugs have the capacity to change the microbiota
by altering the chemical landscape in which the microorganisms live
(e.g., statins, bile acid sequestrants), modulating the host’s ability to
recognize and react to microbes (e.g., immunosuppressants) and/or
directly interfering with the microbiota’s constituents (e.g., antibiotics).
These potential effects have made critical interpretation of microbiota
studies much more difficult. A prominent study that claimed to identify a fecal microbiota signature associated with type 2 diabetes was
later found actually to have identified a signature for patients taking
metformin instead; the effects of this drug on the microbiota were far
greater than the effects of the disease itself. These results highlight the
importance of controlling for clinical variables in microbiota studies.
Antibiotics are the most obvious and best-studied class of drugs
that modulate the microbiota. Multiple groups have demonstrated
that antibiotics exert a considerable effect on the gut microbiota by
depleting antibiotic-sensitive strains. What is more surprising is that
many strains resistant to the antibiotic tested are also eliminated. This
observation highlights the intricate microbe–microbe interactions that
are fundamental to maintenance of the overall microbial community.
For example, treatment with ciprofloxacin, which has little to no activity against clinically relevant anaerobes, leads to a loss of roughly onethird of the bacterial taxa in the gut. This broad effect is likely mediated
by the depletion of certain “keystone” species that are required for the
persistence of other, unrelated species. While many of the observed
antibiotic effects (e.g., loss of specific taxa) are shared across many
different individuals, some effects vary greatly among people. For
example, studies found that microbiota recovery following antibiotic
treatment differed significantly in terms of timing and degree. The
microbiota of most healthy people who received ciprofloxacin for
5 days had completely recovered within 4 weeks, whereas microbiologic changes lasted up to 6 months in other individuals. Moreover,
the degree of variation was compounded by repeated antibiotic
administration, with fewer individuals reverting to their baseline
microbiota after a second course of ciprofloxacin given 6 months after
the first. These findings are consistent with those of microbial ecology
experiments, which also showed that this type of repeated disturbance
leads to less predictable results.
Lifestyle Many seemingly innocuous lifestyle decisions can impact
the human microbiota. For example, a person’s skin and fecal microbiotas are more similar to those of their household members, regardless of
genetic relatedness, than to those of residents of different households.
The degree of similarity in skin microbiotas is even greater if a dog also
lives in the home; in contrast, the presence of a young child does not
accentuate this microbial relatedness. The presumption is that the dog
serves as a more effective “vector” for transmitting microbes during
its frequent direct contact with adults in the household. The type of
setting in which a person lives also impacts the microbiota. Living in a
rural or farm setting leads to a different fecal microbiota than living in
an urban environment. Similarly, the individual’s country of residence
affects the microbiota. An analysis of daily fecal samples from an individual who temporarily (i.e., for a couple of months) moved from the
United States to Thailand demonstrated a large shift in the fecal microbiota that coincided with arrival in Thailand and a reversion in most
respects to the “American” microbial configuration upon return to the
United States. Similarly, immigration to the United States “westernizes”
the microbiome of individuals coming from non-Western countries.
These geography-driven changes probably reflect a combination of
environmental and dietary differences between locations.
Circadian Rhythms Many human biologic processes follow a circadian clock; aspects of physiology are tuned by external cues, including
the degree and timing of ambient light, temperature, and availability of
nutrients. This endogenous biologic clock enables animals to efficiently
adapt to changing environmental conditions. Similarly, the microbiota
maintains a circadian rhythm that is linked to—and helps entrain—the
host’s circadian clock. If circadian oscillations are disrupted in the host,
they are similarly disrupted in the microbiota, and vice versa. These
bacterial vacillations occur at the level of spatial localization within the
intestine, relative species abundance, and bacterial metabolite secretion. Work in the 1960s showed that mice exhibited daily periodicity
of susceptibility to infection with either Streptococcus pneumoniae or
E. coli lipopolysaccharide. Although the fundamental basis for this
difference was not known at the time, it is likely to be related, in part,
to the microbial circadian clock. Derangements of these microbial
oscillations have also been linked to the development of metabolic
diseases and may underlie some of the health hazards associated with
shift work and jet lag.
THE MICROBIOTA AND DISEASE
■ THE HYGIENE HYPOTHESIS
Over the past few decades, abundant epidemiologic data have revealed
an inverse correlation between exposure to microbes and the incidence
of autoimmune and/or atopic diseases (Fig. 471-5). This type of epidemiologic correlation led to the proposal of the “hygiene hypothesis”
in 1989. Initially, this hypothesis focused on the development of atopic
diseases in young children, with the idea that these epidemiologic
observations could “be explained if allergic diseases were prevented by
infection in early childhood, transmitted by unhygienic contact with
older siblings, or acquired prenatally from a mother infected by contact
with her older children.”1
In fact, this notion that differences in living
conditions and environmental exposures contribute to susceptibility to
hay fever (summer catarrh) dates back to at least the early nineteenth
century. The hygiene hypothesis has continued to evolve over the past
three decades and now posits that inadequacies in microbial exposure—
in combination with genetic susceptibilities—lead to a collapse of the
normally highly coordinated, homeostatic immune response. At its
core, the hygiene hypothesis holds that specific early-life microbial
No comments:
Post a Comment
اكتب تعليق حول الموضوع