Vous êtes sur la page 1sur 30

c 




m

Table of Contents
1.m INTRODUCTION............................................................. .......................................2
a.m xOW Px RM CEUTIC  COMP NY DEVEOPS DRUGS.........................4
.m N NOROBOTS: Wx T RE TxEY?..................................................................5
.m PRINCIPES OF N NOROBOTIC PPIC TIONS........................................6
u.m PPIC TIONS OF N NOROBOTS IN MEDIC  FIED...................... ........7
5.1m IMPROVING OXYGEN DEIVERY........................... ............................7
5.1.1 DESIGN ISSUES OF RESPIROCYTE............................................ .8
5.2m xE RT TT CKS........................................... .........................................9
5.2.1m N NOROBOTS IN xE RT SURGERY...................................9
5.2.2m MOECU R SORTING ROTORS...................... ................... 10

5.2.3m PROPEOR............................................................ ..................10


5.2.4m INJECTION OF N NOROBOTS............................ ..................10
5.2.5m N VIG TION............................................................ ................11
5.2.6m POSITIONING............................................................ ................12
5.2.7m DETECTION............................................................... ...............12
5.2.8m DESTRUCTION......................................................... ................12
5.3m N IDE  N NOROBOTIC Px RM CEUTIC  DEIVERY
VExICE..................................................................................................13
5.3.1m COOKING C NCER WITx N NOSxES...........................15
5.3.2m CONTRO OF CE SIGN ING PROCESSES...................18
5.4m REP CING JOINTS WITx BETTER STUFF................................. .....20
5.5m xOW DOES N NOROBOT DETECT CEREBR  NEURYSM?
...................................................................................................................22
£.m M NUF CTURING DESIGN....................... .......................... ............................24
º.m FEW INTERESTING F CTS BOUT N NOROBOTS....................................25

CONCUSION........................................................................ ................................................28

REFERENCES........................................................................ .................................................29


   
m
c  


m

¦ TR¦CT
The path we have chosen is the union of robotics & medicine. The integration of
nanotechnology into medicine is likely to bring some new challenges in medical treatment.
The proposed application of nanorobots can range from common cold to dreadful disease
like cancer. Some such examples can be Pharmacyte, Respirocyte, Microbivores,
Chromallocyte and many more. The study of nanorobots has lead to the field of
Nanomedicine. Nanomedicine offers the prospect of powerful new tools for the treatment of
human diseases and the improvement of human biological systems.

et us peep inside the real world of N¦NOTECHNOLOGY.


dvancement in nanotechnology may allow us to build artificial red blood cells called
Respirocytes capable of carrying oxygen and carbon dioxide molecules (i.e., functions of
natural blood cells). Respirocytes are nanorobots, tiny mechanical devices designed to
operate on the molecular level. Respirocytes can provide a temporary replacement for natural
blood cells in the case of an emergency. Thus Respirocytes will literally change
the treatment of heart disease. One of the most realistic and nearly feasible achievements is
the cure for cancer which is one of the main focuses of this work. From eliminating the side
effects of chemotherapy to treating ¦lzheimer's disease, the potential medical applications
of nanorobots are vast and ambitious. few generations from now someone diagnosed
with cancer will be offered a new alternative to chemotherapy. The traditional treatment of
radiation that kills not just cancer cells but healthy human cells as well, causing hair loss,
fatigue, nausea, depression, and a host of other symptoms. doctor practicing Nanomedicine
would offer the patient an injection of a special type of Nanorobot that would seek out
cancer cells and destroy them, dispelling the disease at the source, leaving healthy cells
untouched. The extent of the hardship to the patient would essentially be a prick to the arm.
person undergoing a nanorobotictreatment could expect to have no awareness of the
molecular devices working inside them, other than rapid betterment of their health.
These nanorobots will be able to repair tissues, clean blood vessels and airways, transform
our physiological capabilities, and even potentially counteract the aging process. The
researchers think their nanorobots could become available around 2015. If this medicine
delivery comes into existence, then there will be no end for our human life. In this paper we
are dealing with nanorobotics.


   
m
c  


m

CH¦PTER 1

INTRODUCTION:

In the 1966 film fantastic voyage, audiences were introduced to their own bodies from the
inside. group of doctors got into a submarine-like ship and were shrunk down to the size of
a cell. They were then injected into a human body to search and destroy a blood clot.
udiences got a fairly realistic look at some of the inner working of the human body.

It¶s a recurrent theme in science fiction: human beings want to live long, healthy, pain-free
lives and we will continue to create (and invest in) ways to do so. Nanotechnology is an
emerging reality that can help us along that path. It won¶t enable humans to shrink , but it
can, however, help us modify and create particles that circulate through the body with as
much control as if we were in there calling the shots.

Nanotechnology is a fascinating science and it offers many challenges. One such challenge
is Nanorobots, which once thought to be a fantasy has come into reality now. Nanorobotics
is the emerging technology field of creating machines or robots whose components are at or
close to the microscopic scale of a nanometer (10í9 meters). More specifically, nanorobotics
refers to the nanotechnology engineering discipline of designing and building nanorobots,
with devices ranging in size from 0.1-10 micrometers and constructed of nanoscale or
molecular components. The names nanobots, nanoids, nanites, nanomachines or
nanomites have also been used to describe these devices currently under research and
development.

The present era of nanotechnology has reached to a stage where programmable and externally
controllable complex machines that are built at molecular level which can work inside the
patient¶s body. Nanotechnology will enable engineers to construct sophisticated nanorobots
that can navigate the human body, transport important molecules, manipulate microscopic
objects and communicate with physicians by way of miniature sensors, motors, manipulators,
power generators and molecular- scale computers. The idea to build a nanorobots comes from
the fact that the body¶s natural nanodevices; the neutrophiles, lymphocytes and white blood
cells constantly rove about the body, repairing damaged tissues, attacking and eating invading
microorganisms, and sweeping up foreign particles for various organ to break down or


   
m
c  


m

excrete. Nanomedical systems designed to perform a specific task with precision at nanoscale
dimension. This allows drugs of nanosize to be used in lower concentration and has an earlier
onset of therapeutic action. It also provides materials for controlled drug delivery by directing
carriers to a specific location. The typical medical nanodevice will probably be a micron-
scale robot assembled from nanoscale parts. These nanorobots can work together in response
to environment stimuli and programmed principles to produce macro scale results.

This chapter explores some unique approaches to delivering drugs and killing cancer that
nanotechnology makes possible. We start with problems pharmaceutical companies have
with drug development and discuss a ³nano-way´ to deliver drugs that addresses this
problem. We then provide a recipe for eliminating cancer and tell you about some new
concepts of artificial blood and bones.


   
m
c  


m

CH¦PTER a

HOW PH¦RM¦CEUTIC¦L COMP¦NY DEVELOP


DRUG
Bioavailability refers to how well a treatment can target specific cells. For the last hundred
years or so, pharmaceutical products have suffered from poor bioavailability their main
approach is to flood the body with drugs that are needed. One aspirin cures a headache but
one hundred aspirins kill the patient. This is especially not good for cancer and
chemotherapy; increasing the amount of toxic drugs eventually kills the patient.

s with any business, companies doing drug development have to choose between risk and
reward. The amount of risk in drug development is incredibly high in terms of both times and
money. The starting point is to determine which molecules of what compounds will be most
effective at curing a disease. Different programs speed up the process but taking a new drug
from research to development to administration still takes a long time. Even today, the
average time between the patent application and marketing of a new medicine is 12 years.
nd a patent expires after only 20 years. To top it off, some research projects are scrapped as
late as advanced stage clinical trials (roughly nine years after kickoff!).

Only 32 new drugs were introduced in 2000 a pretty low figure and the number of new drugs
is increasing. This is not good trade-off, but there may be some light at the end of particular
tunnel: Rapid advances in biotechnological applications can help us in the direction of faster,
less-costly drug development. Nanotechnology, in conjunction with biotechnology, will aid
in more effective medicines while lowering the costs.


   
m
c  


m

CH¦PTER 

N¦NORO OT: WH¦T ¦RE THEY?


Nanorobots are nanodevices that will be used for the purpose of maintaining and protecting
the human body against pathogens. They will have a diameter of about 0.5 to 3 microns and
will be constructed out of parts with dimensions in the range of 1 to 100 nanometers. The
main element used will be carbon in the form of à à


   
 because
of the strength and chemical inertness of these forms. Many other light elements such as
oxygen and nitrogen can be used for special purposes. To avoid being attacked by the host¶s
immune system, the best choice for the exterior coating is a passive diamond coating, The
smoother and more flawless the coating, the less the reaction from the body¶s immune
system. Such devices have been designed in recent years but no working model has been built
so far.

The powering of the nanorobots can be done by metabolising local glucose and oxygen for
energy. In a clinical environment, another option would be externally supplied acoustic
energy. Other sources of energy within the body can also be used to supply the necessary
energy for the devices. They will have simple onboard computers capable of performing
around 1000 or fewer computations per second. This is because their computing needs are
simple. Communication with the device can be achieved by  à 
   
  

navigational network may be installed in the body, with station keeping navigational
elements providing high positional accuracy to all passing nanorobots that interrogate them,
wanting to know their location. This will enable the physician to keep track of the various
devices in the body. These nanorobots will be able to distinguish between different cell types
by checking their surface antigens (they are different for each type of cell). This is
accomplished by the use of chemotactic sensors keyed to the specific antigens on the target
cells.

When the task of the nanorobots is completed, they can be retrieved by allowing them to
exfuse themselves via the usual human excretory channels. They can also be removed by
active scavenger systems. This feature is design-dependent.


  
m
c  


m

CH¦PTER 

PRINCIPLE OF N¦NORO OTIC ¦PPLIC¦TION


The availability of advanced nanomedical instrumentalities should not significantly alter the
classical medical treatment methodology, although the patient experiences and outcomes will
be greatly improved. Treatment in the nanomedical era will become faster and more accurate,
efficient and effective.

Three key required pieces to advance the development and implementation of medical
nanorobotics, according to the paper published by Th
 
        


 h:
1.m Equipment prototyping: Computational nanotechnology provides a key tool for the
fast and effective development of nanorobots, helping in the investigation to address
major aspects on medical instrumentation and device prototyping.

2.m The manufacturing technology: For manufacturing purposes, the nanorobot should be
integrated as a biochip device.

3.m Inside-body transduction: Cell morphology, microbiology, and proteomics are used
as parameters for nanorobot morphology and inside-body interaction. Changes on
chemical gradients and telemetric instrumentation are used for medical prognosis, with
the nanorobots activation based on proteomic over expression.


   !
m
c     
m

TE mm

L T Smm TSm m


E Lm
 EL m

 m
 mEmEL Em

Π 
  
  
 
Œ

Œ
 !"
#  m

The arti i ial mechanical red bl d cell or ³respirocyte´ is a bloodborne spherical 1-micron
diamondoid 1000- atmosphere pressure vessel (Fi 1) with active pumping powered by
endogenous serum glucose, able to deliver 236 times more oxygen to the tissues per unit
volume than natural red cells and to manage carbonic acidity. The nanorobot is made of 18
billion atoms precisely arranged in a diamondoid pressure tank that can be pumped full of up
to 3 billion oxygen (O2) and carbon dioxide (CO2) molecules. Later on, these gases can be
released from the tank in a controlled manner using the same molecular pumps. Respirocytes
mimic the action of the natural hemoglobin-filled red blood cells. Gas concentration sensors
on the outside of each device let the nanorobot know when it is time to load O2 and unload
CO2 (at the lungs), or vice versa (at the tissues). An onboard nanocomputer and numerous
chemical and pressure sensors enable complex device behaviors remotely reprogrammable by
the physician via externally applied acoustic signals.

  
   
m
c  


m

u.1.1 DEIGN IUE OF REPIROCYTE

Figure a respirocyte design

The respirocyte is built of 18 billion precisely arranged structural atoms. It is constructed of


diamondoid honeycomb framework for maximum strength. Thick diamond bulkheads
separate internal tankage volumes. Twelve pumping stations are spaced evenly along an
equatorial circle. Respirocytes exchange gasses via molecular sorting rotors. The rotors have
specially shaped tips to catch particular types of molecules Gas molecules are stored tightly
in tanks. Each respirocyte has three types of rotors. One gathers oxygen at the lungs or in
production before introduction to the body and releases it while traveling through the body.
nother captures carbon dioxide while in the bloodstream and releases it at the lungs. The
third takes in glucose from the bloodstream, which is burned in a reaction similar to cellular
respiration in order to power the respirocyte. nonboard hemomechanical turbine or fuel cell
generates power by combining glucose drawn from the bloodstream and oxygen drawn from
internal storage.. Each power plant develops 0.3 Pico watts of power. One power plant
measures 42 nm x 42 nm x 175 nm in size, constructed of 100 million atoms weighing ~10-
18 kg. Various sensors are needed to acquire the external data essential in regulating gas
input and output operations, tank volume management, and unique protocols. One example is


   "
m
c     
m

constructing a concentration sensor from a sorting rotor. The respirocyte would probably
need only about 1000 operations per second.

5.2 HEARTmTT m

Blood vessels play an important role in supply a blood to all parts of the body. Due to the
fatty deposition on the walls of blood vessels blood will not move freely to all parts of the
body these leads to heart attacks and damage the vital organs.

In general the most common methods of surgery used for heart attacks is

1. By-Pass surgery

2. Angio Plaster

  m TSm mETmS Em

Both of the above methods are risky and number of side effects. As a result patient becomes
very weak. But a surgery-using nanorobot is very simple one. Doctors do their treatment even
without touching the body.

Below figure 3 shows the structure of the nanorobots. It is constructed with various

Π$%

 & 
m

 
   
m
c  


m

nanomechanical devices and nanosensors like.

· Molecular sorting rotors

· Propeller

· Fins

· Sensors

Types of Sensors

1. Chemical sensors -> to find the fatty deposit.

2. Microwave generated sensing -> to generate movement.

3. Chemotactic sensors -> to find cancer cells.

4. coustic sensors-> to guide the nanorobots

u.a.a MOLECUL¦R ORTING ROTOR

It is made up of carbon nanotubes. Simply a sheet of carbon atom forms a carbon nanotube .
roll having only one sheet of carbon atoms thickness is known as single walled carbon
nanotubes (SWNT). Thus the electrical properties of SWNT¶s can be used to generate
mechanical motion from electrical energy. One of the main advantages of these SWNT¶s is,
operating at the molecular level. Nanotube substitutes with nanogears with axle used for
changing the direction of movement.

u.a. PROPELLOR

The word propeller in ship is used to drive forward the device against water. ike that in
nanorobots it is used to drive forward against the blood stream. Fins are fitted along with the
propellers which are used to propel the device. Sensors are fitted externally and internally
with the nanorobots to receive the signal for correct guidance. There are several techniques to
do the heart surgery with the nanorobots. We have to know how to inject nanorobots into our


   
m
c     
m

body, how to move it to the destination place, how to control and remove the device after
surgery.

  m E T mm TSm

We have to find a way to introduce nanorobots into the body for surgery and allowing it to do
the operation without ancillary damage. So nanorobots should be made smaller than the blood
vessels thus making it to travel. Femoral artery in the leg is considered to be the largest artery
in our body. So we inject the nanorobots through this artery.

  m T m

Π'& ( 
  
 
 
( m

Every living thing needs area to move. Like fishes are moved in water, nanorobots use blood
flow for its movement. It must be able to guide the device which makes use of the blood
flow. The devices used for navigation are propeller, fins, jet pump, and electromagnetic
pump. In order to move the nanorobots in blood flow, following things are veryimportant

1. Speed of blood

2. Get through the heart without stuck

3. React with changes in blood flow rate

4. Able to change the direction according to the blood stream

 
   
m
c     
m

To satisfy the above consideration we have to make the nanorobots with electric motors
turning propellers.

  mS T  m

To know the location of nanorobots where it goes we use ultrasonic techni ue. Nanorobots
must be able to produce ultrasonic waves by passing a signal to piezoelectric membrane,
which is in built with the device. Several signals processing techni ues are used to trackthis
ultrasonic signal and finding the location at any time. Instead of ultrasonic wave we use
infrared ray for signal processing.

  mETE T m

To locate a blood clot (or) deposit of arterial plaque we use sensors of different types.
Already preplanned route is available to reach operation site. With the help of preplanned
route we reach the fatty deposited area. (NanoRobots towards a destination) To control the
nanorobots as per our wish, we fit the TV camera in the nanorobots and transmit its picture
outside the body to a remote control unit. Solid-state television camera sensors are used to
receive the signals from the remote station and do the operations according to signals send by
remote control unit. There are pre-programmed microchips available to give appropriate
signals so that nanorobots is initiated externally through a computer.

  mEST T m

The fatty deposits (or) clots are removed using special blades fitted with nanorobots.

Π) " & 


#( 




m

 
   
m
c  


m

Continuous (or) pulse signal is used to activate the blades. These blades physically separate
the deposits from blood vessels. Care should be taken in removing the fatty deposits. Small
deposits of these fatty materials without removing lead to big problem in future.

Production of power is very important for every operation to most efficient one in magnetic
induction. Our body is full of magnetic field. Rotation of nanorobots cuts this magnetic field,
produce power based on  à . To take nanorobots from the body we use two
methods one is retrace our path upstream another is making small surgery to remove.

u.am¦n Ideal Nanorobotic Pharmaceutical Delivery Vehicle

What would an ideal drug delivery vehicle look like? To start with, it would be targetable not
just to specific tissues or organs, but to individual cellular addresses within a tissue or organ.
lternatively, it would be targetable to all individual cells within a given tissue or organ that
possessed a particular characteristic (e.g., all cancer cells, or all bacterial cells of a defined
species). This ideal vehicle would be biocompatible and virtually 100% reliable, with all drug
molecules being delivered only to the desired target cells and none being delivered elsewhere
so that unwanted side effects are eliminated. The ideal vehicle would remain under the
continuous control of the supervising physician, including post-administration. Even after the
vehicles had been injected into the body, the doctor would still be able to activate or
inactivate them remotely, or alter their mode of action or operational parameters. Once
treatment was completed, all of the vehicles could be removed intact from the body, leaving
no lingering evidence of their passage. This hypothetical ideal drug delivery vehicle may be
called a ³Pharmacyte´
Pharmacytes will be self-powered, computer-controlled nanorobotic systems capable of
digitally precise transport, timing, and targeted delivery of pharmaceutical agents to specific
cellular and intracellular destinations within the human body. Drug molecules could be
purposely delivered to one cell, but not to an adjacent cell, in the same tissue. The exemplar
Pharmacyte would not be a relatively passive nanoparticle but rather would be an active
medical nanorobot 1±2 ȝm in size, similar to the respirocyte but slightly larger. It would be
capable of carrying up to ~1 ȝm3 of pharmaceutical payload stored in onboard tanks that are
mechanically offloaded using molecular sorting pumps mounted in the hull, operated under
the control of an onboard nanocomputer. Depending on mission requirements, the payload
could be discharged into the proximate extracellular fluid or delivered directly into the


   
m
c  


m

cytosol using a transmembrane injector mechanism. The sorting pumps are typically
envisioned as ~1,000 nm3-size devices that can transfer ~106 molecules/sec. Each pump
employs reversible binding sites mounted on a rotating structure that cycle between the
interior and exterior of the nanorobot, allowing transport of a specific molecule even against
a considerable concentration gradient.
Other reversible binding sites comprise sensors on the surface of the nanorobot in order to
recognize the unique biochemical signature of specific vascular and cellular addresses,
simultaneously testing encountered biological surfaces for a sufficiently reliable combination
(at least 5±10 in number) of positive-pass and negative-pass molecular markers to ensure
virtually 100% targeting accuracy. Onboard power may be provided by glucose and oxygen
drawn from the local environment (e.g., circulating blood, interstitial fluid, or cytosol) that is
metabolized using fuel cells or other methods for biochemical energy conversion. If needed
for a particular application, deployable mechanical cilia and other locomotive systems can be
added to the Pharmacyte to permit transvascular and transcellular mobility, thus allowing
delivery of pharmaceutical molecules to specific cellular and even intracellular addresses
with negligible error. Because sorting pumps can be operated reversibly, Pharmacytes could
also be used to selectively extract specific molecules from targeted locations as well as
deposit them. Pharmacytes, once depleted of their payloads or having completed their
mission, would be recovered from the patient via centrifuge nanapheresis or by conventional
excretory pathways. The nanorobots might then be recharged, reprogrammed and recycled
for use in a subsequent patient who may need a different pharmaceutical agent targeted to
different tissues or cells than in the first patient.
Phagocytosis and foreign-body granulomatous reaction are major issues for all medical
nanorobots intended to remain in the body for extended durations, though short-duration
Pharmacytes that can quickly be extracted from the body may face somewhat fewer
difficulties. In either case, bloodborne 1 to 2 ȝm Pharmacytes can avoid clearance by the RES
(whether via geometrical trapping or phagocytic uptake) and techniques have been proposed
for phagocyte avoidance and escape at each step in the phagocytic process. Modest
concentrations of Pharmacytes will not embolize small blood vessels because the minimum
viable human capillary that allows passage of intact erythrocytes and white cells is 3 to 4 ȝm
in diameter, which is larger than the largest proposed Pharmacyte. Pharmacytes can also be
equipped with mobility systems to allow mechanically-assisted passage through partially
occluded vessels or unusually narrow spaces such as the interendothelial slits of the spleen.
Targeting ligands or receptors in the cell membrane exterior can be recognized by


   
m
c     
m

chemotactic sensors on the nanorobot surface, but note that the Pharmacyte (as distinguished
from conventional nanoparticles) need not always be endocytosed. For example, in some
cases nanorobots may use transmembrane mechanical nanoinjectors to avoid having to enter
a target cell. Alternatively, if the mission requires cytopenetration then endocytosis of the
nanorobot may be purposely stimulated using biomimetic or completely artificial (including
powered mechanical) methods; after payload delivery, indigestible diamondoid nanorobots
will require exocytosis by similar means. Nanorobot volume is only 1 to 10 ȝm3 compared to
103 to 104 ȝm3 for most human tissue cells so Pharmacytes could be targeted to intracellular
organelles, though nanorobots would have insufficient room to enter one (excepting perhaps
the ER and nucleus) and would have to rely on nanoinjection in those cases.

There are many potential uses of Pharmacytes but it will suffice to briefly mention just two
general classes of applications.
  m  m  Em TmSELLSm

The body is constantly replacing old cells with new ones; the old, damaged cells are
deliberately ³killed´ in a process called apoptosis. Sometimes, however, mutations occur so
that some new cells form when the body doesn¶t need them and old cells don¶t die when they
should ±which, by the way, is a basic definition of cancer cells bypass apoptosis and form a

Π*
#   
m


 
   
m
c     
m

mass of tissue called a tumour.

Pharmacyte is often desired to deliver cytocidal agents to tumor cells. Current methods
involve introducing large quantities of chemotherapy agents into the body in aneffort to kill a
relatively few cancerous cells, with numerous unwanted side effects on healthy cells. Precise
targeting using Pharmacytes can ensure delivery only to the correct cellular addresses, with

Π+  

#
"
 
m

presentation of cytocidal chemical agents literally on a cell-by-cell basis. In one trivial


scenario, the targeted killing of 1 billion cancer cells with each cell capable of being killed by
~106 precisely delivered ~1,000-dalton cytocidal molecules (i.e., lethality similar to bufagin
toxin) would require a total whole-body treatment dose of just ~1015 cytocidal molecules or
~0.001 mm3 (~2 ȝg) of delivered material. This dose could be carried and dispensed by one
trillion Pharmacyte nanorobots (total injected volume of therapeutic nanorobots ~2 cm3)
assuming that only 0.1% of the nanorobots encounter anacceptable target and are allowed to
release a 0.001 ȝm3 cytocidal payload into the targeted cell, while the remaining 99.9% of
the nanorobots release nothing. After initiating cell death, unmetabolized free cytocidal
molecules can be locally reacquired by the Pharmacyte and subsequently transported out of
the patient, thus minimizing any post-treatment collateral damage. Note that the strict size
requirements for macromolecules to reach the leaky vasculature of a tumor and convectively
enter its pores may apply to passively-diffusing payload molecules that might be conveyed
and released by Pharmacytes, but these limits do not apply to the motorized active nanorobots
themselves. Upon arriving in the vicinity of a tumor, the Pharmacyte may deliver its payload
either via direct nanoinjection (for tumor cells adjoining the vasculature) or by progressive

 
   
m
c  


m

cytopenetration through adjacent cells until the targeted tumor cell that awaits payload
delivery is reached.
It is well-known that apoptotic cellular ³death receptors´ can be expressed on both normal
and cancerous cells in the human body, so one challenge for conventional drug-based therapy
is to find some way to activate death receptors selectively on cancer cells only. With
Pharmacytes, such selectivity should be simple and routine using multiple chemosensors, a
benefit that may be characteristic of most future nanorobot-based therapeutics. For example,
if caspase cascade amplification is sufficient to permit single-site activation of the cascade,
then in principle an extracellular nanorobot intending cytocide of a detected cancerous cell
could press onto the outer surface of the target cell an appropriate ligand display tool. This
tool might contain suitably exposed trimeric CD95 (aka Fas) ligand (binds to the
extracellular domains of three CD95 death receptors), TNF or lymphotoxin alpha (binds to
CD120a), po3 ligand aka TWE K (binds to DR3), or po2 ligand. The binding event
would then activate a single death receptor complex, potentially triggering the entire
irreversible cytocidal cascade. If necessary, multiple such display tools could be employed.
This technique avoids much of the storage requirement for bulky consumables aboard the
medical nanorobot. s yet another approach, molecular sorting pumps on the pharmacyte
surface could be used to selectively extract from the cytoplasm of a target cell specific crucial
molecular species of inhibitors of apoptosis (I Ps) that normally hold the apoptotic process
in check. Examples include survivin, commonly found in human cancer cells, the
transcription factor NF-țB, and kt, which delivers a survival signal that inhibits the
apoptosis induced by growth factor withdrawal in neurons, fibroblasts, and lymphoid cells.
Conversely, decoy receptors (DcRs) that compete with DR4 and DR5 for binding to po2
could be saturated with intrinsically harmless but precisely engineered intracellular ³chaff´
ligands. With I Ps removed or DcRs blockaded, apoptosis may be free to proceed.
Pharmacytes could also tag target cells with biochemical substances capable of triggering a
reaction by the body¶s natural defensive or scavenging systems, a strategy called ³phagocytic
flagging´. For example, novel recognition molecules are expressed on the surface of
apoptotic cells. In the case of T lymphocytes, one such molecule is phosphatidylserine, a lipid
that is normally restricted to the inner side of the plasma membrane but, after the induction of
apoptosis, appears on the outside. Cells bearing this molecule on their surface can then be
recognized and removed by phagocytic cells. Seeding the outer wall of a target cell with
phosphatidylserine or other molecules with similar action could activate phagocytic behavior
by natural macrophages that had mistakenly identified the target cell as apoptotic. oading


   #
m
c  


m

the target cell membrane surface with B7 costimulator molecules also permits T-cell
recognition, allowing an immunologic response via the immunological synapse. These
tagging operations should work well against cells that have an apoptotic response that can be
triggered by cytotoxic T cells, such as human cancer cells and cysts.
5.2.2m control of cell signalling processes

second major application area of pharmacytes would be the control of cell Signalling
processes. s a trivial example, Ca++ serves as an intracellular mediator in a wide variety of
cell responses including secretion, cell proliferation, neurotransmission, cellular metabolism
(when complexed to calmodulin), and signal cascade events that are regulated by calcium-
calmodulin-dependent protein kinases and adenylate cyclases. The concentration of free
Ca++ in the extracellular fluid or in the cell¶s internal calcium sequestering compartment
(which is loaded with a binding protein called calsequestrin) is ~10-3 ions/nm3. xowever, in
the cytosol, free Ca++ concentration varies from 6 × 10-8 ions/nm3 for a resting cell up to 3 x
10-6 ions/nm3 when the cell is activated by an extracellular signal; cytosolic levels >10-5
ions/nm3 may be toxic (e.g., via apoptosis). To transmit an artificial Ca++ activation signal to
a typical (20 ȝm)3 tissue cell in ~1 msec, a single pharmacyte stationed in the cytoplasm
must promptly raise the cytosolic ion count from 480,000 Ca++ ions to 24 million Ca++ ions.
This is a transfer rate of ~2.4 × 1010 ions/sec that may be accomplished using ~24,000 hull-
mounted molecular sorting pumps across a total nanorobot emission surface area of ~2.4
ȝm2. Onboard storage volume of ~1 ȝm3 can hold up to ~20 billion calcium atoms, enough
to transmit up to ~1,000 arti- ficial Ca++ signals into the cell even assuming no reabsorption
and recycling of the ions.

Properly configured in cyto pharmacytes could also modify natural intracellular message
traffic according to preprogrammed rules or by following external commands issued by the
supervising physician. In the case of steroids and thyroid hormones, this may involve the
direct manipulation of the signaling molecules themselves (after they have passed through the
cell membrane) or their bound receptor complexes. xowever, most signaling molecules are
absorbed at the cell surface, initiating a signal cascade which may be modulated by
manipulating second-messenger molecules or other components of the in cyto signal cascade.

few basic examples of signal modifying action involving c MP would include:


   "
m
c  


m


¦   single epinephrine molecule received by a beta adrenergic receptor at a cell
surface transduces the activation of dozens of G-protein alpha subunits, each of which in turn
activates a single adenylate cyclase enzyme which cyclizes hundreds of TP molecules into
c MP molecules. The intracellular population of c MP (in muscle or liver target cells) is
normally <10-6Mor ~5 million molecules for a typical (20 ȝm)3 tissue cell. Stimulation by
epinephrine raises the c MP population to ~25 million molecules in a few seconds.
xowever, upon detecting this rising tide of c MP during the first few msec, each in cyto
pharmacyte could quickly amplify this existing chemical signal by releasing 20 million
c MP molecules (occupying a storage volume of ~0.01 ìm3) from onboard inventories in ~1
msec²thus accelerating cellular response time by several orders of magnitude. O
 
Similarly, upon detection of rising c MP levels in target cells, resident pharmacytes could
use molecular pumps to rapidly remove c MP from the cytosol as quickly as it is formed,
even under maximum adrenal stimulation. The diffusion-limited intake current at the basal
concentration (~6 x 10-7 molecules/nm3) for a c MP-absorbing spherical nanodevice 1 ȝm
in radius is ~4 million molecules/ sec, so a single such device could probably keep up with
natural c MP production rates and thus completely extinguish the response by preserving a
flat basal concentration even in the face of a maximum stimulus. ( s a practical matter, it
may be more efficient to control epinephrine generation at its glandular source unless it is
desired to interface with just a single tissue type.) Simultaneously, the c MPabsorbing
nanorobot may hydrolyze the stored c MP in the manner of the c MP phosphodiesterases,
then excrete these deactivated MP messenger molecules back into the cytosol. Similar
methods might be useful in ligand-gated ion channel desensitization or in disease symptom
suppression, as, for example, in suppressing the prolonged elevation of c MP in intestinal
epithelial cells associated with the cholera toxin, which produces severe diarrhea by causing a
large influx of water into the gut.
3  
R   suppression and amplification, an existing chemical signal could be
eliminated and replaced by a different²even an opposite²message pathway using resident
pharmacyte mediators. lternative pathways may be natural or wholly synthetic. Novel
responses to existing signals may be established within the cell to enhance functionality or to
improve stability or controllability. For instance, detection of one species of cytokine by a
pharmacyte could trigger rapid specific absorption of that cytokine and a simultaneous fast
release of another (different) species of cytokine in its place. Such procedures must of course
take into account the many redundant signaling pathways and backup systems (e.g.,


   $
m
c     
m

developmental signals, immune system, blood clotting) that exist within the body. Medical
nanorobots can allow the replacement of many redundant pathways with more refined and
specific responses.
Ô  m    unlinked signal cascades may be artificially linked using in cyto
nanorobots. As a fanciful example, the receipt of epinephrine by pharmacyteslocated in the
capillaries of the brain could trigger these devices to suppress the adrenalin response while
simultaneously releasing chemical messengers producing message cascades that stimulate
production of enkephalins or other opioids, thus encouraging a subjective state of
psychological relaxation rather than the ³fight orflight´ response to certain stressful
conditions.

 mEL m TSm Tm ETTEmSTm

Bones are actually a sort of process: at the cellular level, they undergo constant repair and
rebuilding as they fight gravity, allowing our body to effectively adapt to our environment.
What makes bone to versatile is its basic building material; hydroxyapatite. Osteoblasts
secrete this stuff within a matrix of collagen. The body continuously reabsorbs the
hydroxvapatite crystals and re-deposits new material in their place what process does is
adjust the bone`s thickness in response to changes inthe body `s distribution of weight figure
shows an excellent top ±down hierarchy of strength in the materials that make up bone

Π  



m

comparing actual bone cells to collagen fibers and to individual collagen molecules [ each a
mere3nm in length]m

 
   
m
c     
m

Why is the bone being reabsorbed? Well, the body figures the bone isn¶t needed. Since the
implant is now bearing the greatest amount of load, the body responds by ³pulling back´ on
bone production in that area. Just as the osteoblasts are stimulated into generating more bone
when they¶re confronted by more stresses, they can also be told to back off when the burden
seems to be less. Result: bone generations falls off, and then the body starts to reabsorb

Π  # 
m

existing bone. The problem is how to keep the bone in the business of rebuilding itself.

Looking specifically at hip implants, another source of contention is the plastic that is used in
the socket portion. A traditional hip implant has a metal ball and a plastic socket, as shown in
fig. the metal rubs against the plastic socket, plastic particles break off, and the body responds
to foreign fragments with inflammation, resulting in bone loss at the femur. Researchers at
the Tokyo University are developing a biocompatible polymer joint that resembles
phospholipids. Using such a polymer joint fools the body into not attacking the residual
flakes; no inflammatory response means no weakening of the bone.

Bioactive materials have the ability to interact with living tissue and are the most promising
approach to ensuring a strong, long lasting adhesive interface between the implant and the
surrounding tissue. Now they have come up with a process that involves injecting the
affected joint with a soft material that then hardens at the molecular level, bondinga damaged
bone together. More specifically, the material reacts to body fluids to form apatite, the body¶s

 
   
m
c     
m

own ceramic. The resulting material is better than the existing artificial bone material; it¶s
strong and hard as actual bone. Such a material is considered a ³bioactive ceramic´ the body
activates the material and helps form it into a ceramic. The approach combines various
sophisticated understandings of materials, chemistry and biology at the molecular level,
which is what nanotechnology, is all about.

 mmESmm TmETE Tmm EE Lm


ES
mm
A ?  l or i m   is a cerebrovascular disorder in which weakness in the wall
of a cerebral artery or vein causes a localizeddilation or ballooning of the blood vessel.
Aneurysms may result from congenital defects, preexisting conditions such as high blood
pressure and atherosclerosis (the buildup of fatty deposits in the arteries), or head trauma.
Cerebral aneurysms occur more commonly in adults than in children but they may occur at
any age.
  
m m
m
?
mim :
 the nanorobots enter the vessel and flow with the bloodstream  the nanorobots are

Π,& 



  #m

moving through the vessel with the fluid ? the aneurysm saccular bulb begins to become
visible at the vessel wall nanorobots move closer to the vessel deformation mixed with
the plasma, NOS (nitric oxide synthase) signals can be detected as the chemical gradient
changes, denoting proteomic overexpression  the same workspace viewed without red

 
   
m
c  


m

cells g) the nanobiosensor is activated as the nanorobots move closer to the aneurysm,
emitting RF signals sent to the cell phoneh) as the nanorobots keep flowing, the chemical
signals become weaker, deactivating the nanorobot transmission i) red cells and nanorobots
flow with the bloodstream until they leave the vessel.
Finally, here¶s a key excerpt from the conclusion and outlook. ³The nanomachine platform
design was based on clinical data, proteomic signals, cell morphology, and numerical
analysis. For the proposed model, the nanorobots were able to recognize chemical gradient
changes in the bloodstream, retrieving information about the position inside the vessel as
intracranial aneurysm detection. n important and interesting aspect in the current
development is the fact that this platform, presented in terms of device prototyping and
system architecture integration, can also be useful for a broad range of applications in
medicine.´


   
m
c  


m

CH¦PTER £:

M¦NUF¦CTURING DEIGN:

The approach taken in our development is called nanobhis (Nano-Build xardware Integrated
System). It represents a joint set of well-established techniques and new methodologies from
nanotechnology with the aim of manufacturing nanorobots. The nanorobot must be equipped
with the necessary devices for monitoring the most important aspects of its operational
workspace. Depending on the case the temperature, Concentration of chemicals in the water,
and electrical conductivity, are some of relevant parameters when monitoring hydrological
resources. Geographically distributed teams of nanorobots are expected to open new
possibilities on agricultural and environmental applications with a larger spectrum of details
not seen whenever. For such aims, computing processing, energy supply, and data
transmission capabilities can be addressed through embedded integrated circuits, using
advances on technologies derived from VSI design. CMOS VSI design using deep
ultraviolet lithography provides high precision and a commercial massively way for
manufacturing nanodevices and nanoelectronics. The CMOS industry may thrive successfully
the pathway to enable nanorobots assembly, where the jointly use of nanophotonic and
nanotubes may even accelerate further the actual levels of resolution ranging from 248nm to
157nm devices. To validate designs and to achieve a successful implementation, the use of
VxD has become the most common methodology utilized in the industry of integrated
circuits.

CH¦PTER º:


   
m
c  


m

FEW INTERETING F¦CT ¦ OUT N¦NORO OT:

· What chemical elements would medical nanorobots be made of?

The typical medical nanodevice is made up of micronscale robot assembled from nanoscale
parts. These parts could range in size from 1-100 nm (1 nm = 10-9 meter), and might be fitted
together to make a working machine measuring perhaps 0.5-3 microns (1 micron = 10-6
meter) in diameter. Three microns is about the maximum size for blood borne medical
nanorobots, due to the capillary passage requirement.

· Could human body fluids get inside the nanorobot?

From a medical standpoint, it makes sense to regard the nanorobot as having two spaces,
which should be considered separately its interior and its exterior. It is true that the nanorobot
exterior will be exposed to the diverse chemical brew that makes up our human biochemistry.
But the interior of the nanorobot may be a highly controlled environment, possibly a vacuum,
into which external liquids cannot normally intrude. Of course it may often be necessary for a
nanorobot to import external fluids in a controlled manner for chemical analysis or other
purposes. But the important thing is that the device will be watertight and airtight. Body
fluids cannot get into the interior of the device, unless these fluids are purposely pumped in
for some specific reason.

· How would the nanorobots be retrieved from the body?

Some nanodevices4 will be able to exfuse themselves from the body via the usual human
excretory channels; others will be designed to allow ready exfusion by medical personnel
using apheresis-like processes (commonly called nanapheresis) or active scavenger systems.
It is very design dependent. In the case of the respirocytes, the removal procedure is fairly
simple: "Once a therapeutic purpose is completed, it may be desirable to extract artificial
devices from circulation. Onboard water ballast control is extremely useful during respirocyte
exfusion from the blood. Blood to be cleared may be passed from the patient to a specialized
centrifugation apparatus where acoustic transmitters command respirocytes to establish
neutral buoyancy. No other solid blood component can maintain exact neutral buoyancy,
hence those other components precipitate outward during gentle centrifugation and are drawn
off and added back to filtered plasma on the other side of the apparatus. Meanwhile, after a


   
m
c  


m

period of centrifugation, the plasma, containing mostly suspended respirocytes but few other
solids, is drawn off through a 1-micron filter, removing the respirocytes. Filtered plasma is
recombined with centrifuged solid components and returned undamaged to the patient's body.
The rate of separation is further enhanced either by commanding respirocytes to empty all
tanks, lowering net density to 66% of blood plasma density, or by commanding respirocytes
to blow a 5-micron O2 gas bubble to which the device may adhere via surface tension,
allowing it to rise at 45 mm/hour under normal gravitational acceleration."

· How fast can medical nanorobots replicate inside the human body?

This is a very common error. Medical nanorobots need not EVER replicate. In fact, it is
unlikely that the FD (or its future equivalent) would ever approve for general use a medical
nanodevice that was capable of    replication. Except in the most unusual of
circumstances, you would never want anything that could replicate itself to be turned loose
inside your body. Replicating bacteria are trouble enough!

Replication is a crucial basic capability for molecular manufacturing. But aside from the most
aggressive applications, there is simply no good reason to risk manufacturing "fertile"
nanorobots inside the human body, when "mule" nanorobots can be manufactured so cheaply,
conveniently, and in such vast numbers outside of the human body. Replicators will almost
certainly be very tightly regulated by governments everywhere.

· Will medical nanorobots possess a humanlike artificial intelligence?

This is another common error. Many medical nanorobots will have very simple computers on
board each device. Respirocytes, for example, have only a ~1,000 operations/sec computer on
board each device far less computing power.

· How would you communicate with the machines as they do their work?

One of the simplest ways to send broadcast-type messages into the body, to be received by 
  nanorobots, is acoustic messaging. device similar to an ultrasound probe would
encode messages on acoustic carrier waves at frequencies between 1-10 Mxz. Thus the
supervising physician can easily send new commands or parameters to nanorobots already at
work inside the body. Each nanorobot has its own power supply, computer, and sensorium,


   !
m
c  


m

thus can receive the physician's messages via acoustic sensors, then compute and implement
the appropriate response. The other half of the process is getting messages back out of the
body, from the working nanodevices out to the physician. This can also be done acoustically.
xowever, onboard power requirements for micron-scale acoustic wave generators in water
dictate a maximum practical transmission range of at most a few hundred microns for each
individual nanorobot. Therefore it is convenient to establish an internal communications
network that can collect local messages and pass them along to a central location, which the
physician can then monitor using sensitive ultrasound detectors to receive the messages. Such
a network can probably be deployed inside a patient in less than an hour, may involve up to
100 billion mobile nanorobotic network nodes, and may release at most 60 watts of waste
heat (less than the 100-watt human body basal rate) assuming a (worst case) full 100%
network duty cycle.

· What form of detection system would medical nanorobots use to


distinguish between differing cell types?

Each cell type has its own unique set of surface antigens. Other cell surface antigens indicate
the health status of the cell, the parent organ type, the species of the animal, and even the
identity of the individuala kind of biochemical Social Security Number.

Example: One very simple nanorobot that I designed a few years ago is the artificial
mechanical red cell, which I call a "respirocyte". The Respirocyte measures about 1 micron in
diameter and just floats along in the bloodstream. It is a spherical nanorobot made of 18
billion atoms. These atoms are mostly carbon atoms arranged as diamond in a porous lattice
structure inside the spherical shell. The respirocyte is essentially a tiny pressure tank that can
be pumped full of up to 9 billion oxygen (O2) and carbon dioxide (CO2) molecules. ater on,
these gases can be released from the tiny tank in a controlled manner. The gases are stored
onboard at pressures up to about 1000 atmospheres. (Respirocytes can be rendered
completely non-flammable by constructing the device internally of sapphire, a flameproof
material with chemical and mechanical properties otherwise similar to diamond.)

CONCLUION:


   #
m
c  


m

Nanotechnology provides the potential for reverse aging, curing physical diseases,
manufacture consumer goods at molecular level. s we have seen the wide application of
nanorobots in field of medicine, its advantages and usability makes it evolving technology in
the coming future. Not only in medicine but the magic of nanotechnology has spread in
various fields like information and communications, food resources, consumer goods,
chemistry and environment.

Nature has created nanostructures for billenia. Biological systems are an existing proof of
molecular nanotechnology. Rather than keep our eyes fixed on the far future, let us start now
by creating some actual working devices that will allow us to cure some of the most deadly
ailments known, as well as advance our capabilities directly, rather than as the side effects of
other technologies. There will be a day when eliminating cancer cells are mere an out patient
medical procedure.

REFERENCE:


   "
m
c  


m

1.m Nanorobotics as medicament: (Perfect solution for cancer): Emerging Trends


in Robotics and Communication Technologies (INTER CT), 2010
International Conference. http://ieeexplore.ieee.org, IEEE CONFERENCES

2.m Use of nanorobots in heart transplantation: Emerging Trends in Robotics and


Communication Technologies (INTER CT), 2010 International Conference.
http://ieeexplore.ieee.org, IEEE CONFERENCES

3.m Nanorobots in cancer treatment: Emerging Trends in Robotics and


Communication Technologies (INTER CT), 2010 International Conference
on , http://ieeexplore.ieee.org, IEEE CONFERENCES

4.m Nanotechnology: Book by Richard booker and earl boysen , wiley


publications US , 2010 edition.

5.m Nanotechnology : a gentle introduction to the next big idea by Mark .


Ratner, Daniel Ratner

6.m Introduction to Nanotechnology by Charles P. Poole, Jr., Charles P. Poole,


Frank J. Owens

7.m Cancer-fighting technology (http:/ / www. physorg. com/ news116071209.


html)


   $
m

Vous aimerez peut-être aussi